WO2017101796A1 - 酞嗪酮衍生物、其制备方法及用途 - Google Patents

酞嗪酮衍生物、其制备方法及用途 Download PDF

Info

Publication number
WO2017101796A1
WO2017101796A1 PCT/CN2016/110007 CN2016110007W WO2017101796A1 WO 2017101796 A1 WO2017101796 A1 WO 2017101796A1 CN 2016110007 W CN2016110007 W CN 2016110007W WO 2017101796 A1 WO2017101796 A1 WO 2017101796A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
group
dihydro
membered
pyrazine
Prior art date
Application number
PCT/CN2016/110007
Other languages
English (en)
French (fr)
Inventor
刘钢
于华
任云
杜静
杨定菊
李晓勇
王坤建
刘伟
唐建川
吴勇勇
曾宏
卿燕
宋宏梅
李少华
葛勇
王利春
王晶翼
Original Assignee
四川科伦博泰生物医药股份有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 四川科伦博泰生物医药股份有限公司 filed Critical 四川科伦博泰生物医药股份有限公司
Priority to CN201680007644.0A priority Critical patent/CN107207504B/zh
Publication of WO2017101796A1 publication Critical patent/WO2017101796A1/zh

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/502Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with carbocyclic ring systems, e.g. cinnoline, phthalazine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains three hetero rings
    • C07D471/14Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • the present application relates to the field of medicine, and in particular to a pyridazinone derivative, a preparation method thereof and use thereof.
  • PARP Poly ADP-Ribose Polymerase
  • ADP is used as a substrate to synthesize poly(ARP) ribose (PRA) on receptor proteins (including itself) (Sakamoto- Hojo E T, Balajee A S. Targeting Poly (ADP) ribose Polymerase I (PARP-I) and PARP-I interacting Proteins for cancer treatment.
  • ARP poly(ARP) ribose
  • PRA poly(ARP) ribose
  • PARP-I ribose Polymerase I
  • PARP-I PARP-I interacting Proteins for cancer treatment. Anticancer Agents Med Chem, 2008, 8(4): 402-416).
  • PARP-1 is involved in DNA damage repair and transcriptional regulation and is considered to be an important regulator of cell survival and death. It is also involved in the regulation of some transcription factors in tumorigenesis and inflammation (Peralta-Leal A, Rodriguez-Vargas J M, Aguilar-Quesada R, et al. PARP inhibitors: New partners in the therapy of cancer and inflammatory diseases. Free Radical Biol Med, 2009, 47(l): 13-26). Up to now, PARP-1 has been found to be highly expressed in various human malignant tumors, such as malignant lymphoma, breast cancer, Ewing's sarcoma, and hepatocellular carcinoma.
  • PARP-1 activity inhibitor can promote cancer cell death.
  • a large number of studies have confirmed that drug inhibition or gene knockout PARP-1 can not only avoid tissue damage caused by oxidative stress-related diseases, but also improve the prognosis of cancer patients.
  • the use of PARP-1 inhibitor alone also has a killing effect on tumors (mainly breast cancer) that are defective in DNA damage repair.
  • the literature also reports the relationship between PARP-1 inhibitors and angiogenesis.
  • VEGF vascular endothelial growth factor
  • the inventors of the present application have systematically studied the structure-activity relationship of pyridazinone compounds and found a class of compounds having excellent PARP inhibitory activity, and the present application has been completed based on the above findings.
  • a first aspect of the application provides a compound of formula I, a prodrug, metabolite form, pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystalline form thereof,
  • a and B together with the attached atoms form an alicyclic, heteroalicyclic, aromatic or heteroaryl ring; optionally wherein the alicyclic, heteroalicyclic, aromatic or heteroaryl ring is independently one or more One (for example 1, 2, 3 or 4) selected from the group consisting of halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aromatic Base, heteroaryl, RO-, ROC(O)-, RC(O)O-, RC(O)-, RR'N-, RC(O)N(R")-, RR'NC(O) - substituted with a substituent of RS(O) a - wherein a is 0, 1 or 2;
  • D and E are each independently selected from C and N, and together with the attached atoms form a 5-10 membered ring X, wherein said ring X is an alicyclic, alicyclic, aromatic or heteroaryl ring; preferably, Ring X is a nitrogen-containing aliphatic heterocyclic or heteroaryl ring; preferably, the ring X is a five- or six-membered nitrogen-containing heterocyclic or heteroaryl ring; preferably, the ring X is five or six a nitrogen-containing heteroaromatic ring; the ring X is, for example, a pyrrole ring, an imidazole ring, a pyrazole ring, a triazole ring, a pyridone ring or a piperidine ring;
  • R 1 , R 2 , R 3 and R 4 are each independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, alkenyl, alkynyl, cycloalkyl, heteroheterocyclyl, Aryl, heteroaryl, RO-, ROC(O)-, RC(O)O-, RC(O)-, RR'N-, RS(O) a -, RC(O)N(R") And RR 'NC(O)-; optionally wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heteroheterocyclyl, aryl or heteroaryl are each independently one or more ( For example, 1, 2, 3 or 4) are selected from the group consisting of halogen, hydroxy, amino, cyano, carboxy, nitro, C 1-4 alkyl, C 1-4 alkoxy, 3-8 membered cycloalkyl, 5
  • R 5 and R 6 are each independently selected from the group consisting of hydrogen, oxy, halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, alkenyl, alkynyl, cycloalkyl, heteroheterocyclyl, Aryl, heteroaryl, RO-, RC(O)-, ROC(O)-, RC(O)O-, RR'N-, RC(O)N(R")-, RR'NC(O )-, ROC(O)N(R')-, RR'NC(O)N(R")-, RS(O) a -, RR'NSO 2 - and RS(O) 2 N(R')
  • the alkyl, alkenyl, alkynyl, cycloalkyl, heteroheterocyclyl, aryl or heteroaryl group are each independently one or more (eg 1, 2, 3 or 4) a) selected from the group consisting of halogen,
  • R 5 or R 6 and form a polycyclic ring system on the ring X to which it is attached, wherein said R 5 or R 6 is selected from the group consisting of a 3-8 membered alicyclic ring, a 5-8 membered aliphatic heterocyclic ring, and a 6-10 membered aromatic group. Ring and 5-10 membered heteroaryl ring;
  • R, R' and R" are each independently selected from the group consisting of hydrogen, C 1-10 alkyl, 3-20 membered cycloalkyl, 5-20 membered heteroheterocyclyl, 6-20 membered aryl and 5-20 membered hetero An aryl group, optionally wherein the C 1-10 alkyl group, the 3-20 membered cycloalkyl group, the 5-20 membered alicyclic group, the 6-20 membered aryl group or the 5-20 membered heteroaryl group are each independently One or more (eg 1, 2, 3 or 4) ground molybdenum is selected from the group consisting of halogen, hydroxy, amino, cyano, carboxy, nitro, C 1-4 alkyl, halo C 1-4 alkyl, hydroxy C 1-4 alkyl, C 1-4 alkoxy, 3-8 membered cycloalkyl, 5-8 membered heteroheterocyclyl, 6-10 membered aryl, 5-10 membered heteroary
  • the limiting condition of the compound of Formula I is
  • R 5 is not H, hydroxy, methyl, isopropyl, cyclopropyl, trifluoromethyl, difluoromethyl and trifluoroethyl;
  • the ring X is not pyrrolidine, 2-pyrrolidone, morpholine, 3-morpholinone, tetrahydrofuran, 2-oxazolidinone, tetrahydrothiophene or thiophene sulfone;
  • a and B together with the attached atoms form an aromatic ring (eg, a 6-20 membered aromatic ring, a 6-14 membered aromatic ring, or a 6-10 membered aromatic ring).
  • the aromatic ring is unsubstituted or each independently one or more (eg 1, 2, 3 or 4) selected from halogen (eg fluorine, chlorine, bromine or iodine), hydroxyl, amino, cyanide
  • halogen eg fluorine, chlorine, bromine or iodine
  • a and B together with the attached atoms, form a phenyl ring in the compound of Formula I, wherein the phenyl ring is unsubstituted or independently one or more (eg, 1 , 2, 3 or 4) substituents selected from halogen (e.g., fluorine).
  • halogen e.g., fluorine
  • the compounds R 1, R 2, R 3 and R 4 are each independently selected from hydrogen, halogen, hydroxy, amino, cyano, carboxy, nitro, C of the formula I 1-10 alkyl and halogenated C 1-10 alkyl.
  • R 1 , R 2 , R 3 and R 4 are each independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, cyano, C 1-6 alkyl. (e.g., methyl) and halo C 1-6 alkyl (e.g., trifluoromethyl).
  • R 1 , R 2 and R 3 are a hydrogen atom.
  • R 4 in the compound of Formula I is a halogen.
  • the compound of the formula I R 4 is fluoro.
  • R 5 and R 6 are each independently selected from the group consisting of non-existing, hydrogen, oxy, halogen, C 1-10 alkyl, 3-20-membered naphthenic Base, 3-20 membered heteroheterocyclyl, 5-10 membered heteroaryl, RO-, RC(O)-, ROC(O)-, RR'N-, RR'NC(O)-, RS(O a - and RR'NSO 2 -; optionally wherein the C 1-10 alkyl group, the 3-20 membered cycloalkyl group, the 3-20 membered alicyclic group or the 5-10 membered heteroaryl group are each independently substituted with one or more (e.g.
  • halogen preferably fluoro
  • R and R' are each independently selected from the group consisting of hydrogen, C 1-6 alkyl, 3-8 membered cycloalkyl, 5-8 membered heteroheterocyclyl, 6-10 membered aryl, and 5-10 membered heteroaryl;
  • the C 1-6 alkyl group, the 3-8 membered cycloalkyl group, the 5-8 membered alicyclic group, the 6-10 membered aryl group, the 5-10 membered heteroaryl group are each independently one Or a plurality (for example 1, 2, 3 or 4) selected from the group consisting of halogen, hydroxy, amino, cyano, C 1-4 alkyl, halo C 1-4 alkyl, hydroxy C 1-4 alkyl, RS Substituent substitution of (O) 2 -, C 1-4 alkoxy, 3-8 membered cycloalkyl, 5-8 membered alicyclic, 6-10 membered aryl and 5-10 membered heteroaryl;
  • R 5 or R 6 and forming a polycyclic ring system on the ring X to which it is attached, wherein said R 5 or R 6 is selected from the group consisting of an aliphatic heterocyclic ring (for example, a 5-8-membered heterocyclic ring) and a heteroaromatic ring (for example, 5) a -10 membered heteroaryl ring); preferably, the alicyclic or heteroaryl ring is a 5- or 6-membered nitrogen-containing heterocyclic or heteroaryl ring; for example, a pyrrolidine ring, a pyrrole ring, an imidazole ring, a pyrazole Ring, triazole ring, oxazole ring, thiazole ring, piperidine ring, piperazine ring, pyrazine ring, morpholine ring, thiomorpholine ring, pyridine ring, pyrimidine ring or pyridazine ring.
  • R 5 and R 6 are each independently selected from the group consisting of absent, hydrogen, oxy, halogen (eg, F, Cl, Br, or I), C 1- 6 alkyl and substituted C 1-6 alkyl (eg methyl, ethyl, fluoromethyl, difluoromethyl, trifluoromethyl, 1,1-difluoroethyl, cyclopropyl-difluoro Methyl, hydroxymethyl, 1-hydroxyethyl, 2-hydroxyethyl, 1-aminoethyl, cyclopropylmethyl, 2-hydroxy-2-propyl), C1-6 alkoxy (eg Methoxy, ethoxy, propoxy, tert-butoxy, cyclopropyloxy,), 3-8 membered cycloalkyl (eg cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl), Substitute
  • R 5 is selected from the group consisting of hydrogen, oxy, halogen, C 1-10 alkyl, 3-20 membered cycloalkyl, 3-20 membered alicyclic.
  • C 1-10 alkyl group, the 3-20 membered cycloalkyl group, the 3-20 membered heteroheterocyclyl group or the 5-10 membered heteroaryl group are each independently one or more (eg, 1 , 2, 3 or 4) selected from halogen (preferably fluorine), hydroxyl, amino, cyano, C 1-6 alkyl, C 1-6 alkoxy, 3-8 membered cycloalkyl, 3-8 Substituted by a substituent of a monoaliphatic heterocyclic group, a 6-10 membered aryl group and a 5-10 membered heteroaryl group, wherein a is 0, 1 or 2;
  • R 5 and forming a polycyclic ring system on the ring X to which it is attached, wherein R 5 is selected from the group consisting of an aliphatic heterocyclic ring (for example, a 5-8-membered heterocyclic ring) and a heteroaromatic ring (for example, a 5-10 membered heteroaryl ring)
  • the heterocyclic or heteroaryl ring is a five- or six-membered nitrogen-containing heteroalicyclic or heteroaryl ring; for example, a pyrrolidine ring, a pyrrole ring, an imidazole ring, a pyrazole ring, a triazole ring , an oxazole ring, a thiazole ring, a piperidine ring, a piperazine ring, a pyrazine ring, a morpholine ring, a thiomorpholine ring, a pyridine ring, a pyrimidine
  • the R 6 is absent or selected from the group consisting of hydrogen, oxy, halogen, C 1-10 alkyl, 3-20 membered cycloalkyl, 3-20 membered heteroheterocyclyl, 5-10 membered heteroaryl, RO- , RC(O)-, ROC(O)-, RR'N-, RR'NC(O)-, RS(O) a - and RR'NSO 2 -; optionally, wherein said C 1-10
  • the alkyl group, the 3-20 membered cycloalkyl group, the 3-20 membered alicyclic group or the 5-10 membered heteroaryl group are each independently selected from one or more (e.g., 1, 2, 3 or 4) halogens ( Preferred is fluoro), hydroxy, amino, cyano, C 1-6 alkyl, C 1-6 alkoxy, 3-8 membered cycloalkyl, 3-8 membered heteroheterocyclyl, 6-10 membered
  • R and R' are each independently selected from the group consisting of hydrogen, C 1-6 alkyl, 3-8 membered cycloalkyl, 5-8 membered heteroheterocyclyl, 6-10 membered aryl, and 5-10 membered heteroaryl;
  • the C 1-6 alkyl group, the 3-8 membered cycloalkyl group, the 5-8 membered alicyclic group, the 6-10 membered aryl group, the 5-10 membered heteroaryl group are each independently one Or a plurality (for example 1, 2, 3 or 4) selected from the group consisting of halogen, hydroxy, amino, cyano, C 1-4 alkyl, halo C 1-4 alkyl, hydroxy C 1-4 alkyl, RS Substituent substitution of (O) 2 -, C 1-4 alkoxy, 3-8 membered cycloalkyl, 5-8 membered alicyclic, 6-10 membered aryl and 5-10 membered heteroaryl,
  • R 5 is selected from the group consisting of hydrogen, oxy, halogen, C 1-10 alkyl, 3-20 membered cycloalkyl, 3-20 membered alicyclic.
  • R and R' are each independently selected from the group consisting of hydrogen, C 1-6 alkyl and 3-8 membered cycloalkyl; optionally wherein the C 1-6 alkyl group and the 3-8 membered cycloalkyl group are each Independently one or more (eg 1, 2, 3 or 4) selected from the group consisting of halogen, hydroxy, amino, cyano, C 1-4 alkyl, C 1-4 alkoxy and C 1-4 alkyl The substituent of the sulfonyl group is substituted.
  • R 5 is selected from the group consisting of hydrogen, oxy, halo, C 1-6 alkyl, 3-8 membered cycloalkyl, 3-8 alicyclic heterocycle.
  • C 1-6 alkyl group, the 3-8 membered cycloalkyl group, the 3-8 membered heteroheterocyclyl group, and the 5-6 membered heteroaryl group are each independently one or more (eg, 1 , 2, 3 or 4) substituents selected from the group consisting of halogen (preferably fluorine), hydroxyl, amino, cyano, C 1-4 alkyl, C 1-4 alkoxy and 3-6 membered cycloalkyl Where a is 0, 1 or 2;
  • R and R' are each independently selected from the group consisting of hydrogen, C 1-4 alkyl and 3-6 membered cycloalkyl; optionally wherein the C 1-4 alkyl group and the 3-6 membered cycloalkyl group are each Independently one or more (eg 1, 2, 3 or 4) selected from the group consisting of halogen, hydroxy, amino, cyano, C 1-2 alkyl, C 1-2 alkoxy and C 1-2 alkyl The substituent of the sulfonyl group is substituted.
  • R 5 is selected from the group consisting of hydrogen, oxy, halo, C 1-4 alkyl, 3-6 membered cycloalkyl, 3-6 alicyclic heterocycle.
  • C 1-4 alkyl group, the 3-6 membered cycloalkyl group, the 3-6 membered alicyclic group, the 5-6 membered heteroaryl group are each independently one or more (eg, 1 , 2, 3 or 4) substituents selected from the group consisting of halogen (preferably fluorine), hydroxyl, amino, cyano, C 1-2 alkyl, C 1-2 alkoxy and 3-6 membered cycloalkyl Where a is 0, 1 or 2;
  • R and R' are each independently selected from the group consisting of hydrogen, C 1-2 alkyl and 3-6 membered cycloalkyl; optionally wherein the C 1-2 alkyl group and the 3-6 membered cycloalkyl group are each Independently one or more (eg 1, 2, 3 or 4) selected from the group consisting of halogen, hydroxy, amino, cyano, C 1-2 alkyl, C 1-2 alkoxy and C 1-2 alkyl The substituent of the sulfonyl group is substituted.
  • R 5 is selected from the group consisting of hydrogen, bromo, hydroxymethyl, 1-hydroxyethyl, 2-hydroxy-2-propyl, difluoromethyl, tri Fluoromethyl, 1,1-difluoroethyl, pentafluoroethyl, cyclopropyl-difluoromethyl, 1-aminoethyl, cyclopropylmethyl, cyclopropyl, 1-methyl-4-pyridyl Azyl, 1-methyl-3-pyrazolyl, 2-methyl-[1.3.4]oxadiazol-5-yl, 3-methyl-[1.2.4]oxodiazole-5- Base, methoxy, cyclopropyloxy, acetyl, ethoxycarbonyl, cyclopropylamino, aminoacyl, carbamoyl, N,N-dimethylamino, ethoxyyl, 2-hydroxyethylamine Acyl, 2-
  • R 6 is absent or selected from the group consisting of hydrogen and a halogenated C 1-10 alkyl group (for example, a halogenated C 1-6 alkyl group; for example, a halogenated C 1 ) -4 alkyl; for example trifluoromethyl).
  • a halogenated C 1-10 alkyl group for example, a halogenated C 1-6 alkyl group; for example, a halogenated C 1 ) -4 alkyl; for example trifluoromethyl.
  • the compound of the formula I R 6 is absent or selected from hydrogen and trifluoromethyl.
  • the compound has a structure as shown in Formula I-A.
  • a and B together with the attached atoms form an alicyclic, heteroalicyclic, aromatic or heteroaryl ring, optionally wherein the alicyclic, heteroalicyclic, aromatic or heteroaryl ring is independently one or more One (for example 1, 2, 3 or 4) selected from the group consisting of halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aromatic Base, heteroaryl, RO-, ROC(O)-, RC(O)O-, RC(O)-, RR'N-, RC(O)N(R")-, RR'NC(O) Substituting a substituent with RS(O) a wherein a is 0, 1 or 2;
  • Ring X is an alicyclic, aliphatic heterocyclic ring, aromatic ring or heteroaryl ring, preferably, said ring X is a nitrogen-containing aliphatic heterocyclic or heteroaryl ring; preferably, said ring X is a five- or six-membered nitrogen-containing ring An alicyclic or heteroaromatic ring; preferably, the ring X is a five- or six-membered nitrogen-containing heteroaryl ring; the ring X is, for example, a pyrrole ring, an imidazole ring, a pyrazole ring, a triazole ring or a piperidine ring;
  • R 1 , R 2 , R 3 and R 4 are each independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, alkenyl, alkynyl, cycloalkyl, heteroheterocyclyl, Aryl, heteroaryl, RO-, ROC(O)-, RC(O)O-, RC(O)-, RR'N-, RS(O) a -, RC(O)N(R') And RR 'NC(O)-; optionally wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heteroheterocyclyl, aryl, heteroaryl are each independently one or more ( For example, 1, 2, 3 or 4) are selected from the group consisting of halogen, hydroxy, amino, cyano, carboxy, nitro, C 1-4 alkyl, C 1-4 alkoxy, 3-8 membered cycloalkyl,
  • R 5 and R 6 are each independently selected from the group consisting of hydrogen, oxy, halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, alkenyl, alkynyl, cycloalkyl, heteroheterocyclyl, Aryl, heteroaryl, RO-, RC(O)-, ROC(O)-, RC(O)O-, RR'N-, RC(O)N(R")-, RR'NC(O )-, ROC(O)N(R')-, RR'NC(O)N(R")-, RS(O) a -, RR'NSO 2 - and RS(O) 2 N(R')
  • the alkyl, alkenyl, alkynyl, cycloalkyl, heteroheterocyclyl, aryl or heteroaryl group are each independently one or more (eg 1, 2, 3 or 4) a) selected from the group consisting of halogen,
  • R 5 or R 6 and forming a polycyclic ring system on the ring X to which it is attached, wherein said R 5 or R 6 is selected from the group consisting of a 3-8 membered alicyclic ring, a 5-8 membered aliphatic heterocyclic ring, and a 6-10 membered aromatic group. Ring and 5-10 membered heteroaryl ring;
  • R, R' and R" are each independently selected from the group consisting of hydrogen, C 1-10 alkyl, 3-20 membered cycloalkyl, 5-20 membered heteroheterocyclyl, 6-20 membered aryl and 5-20 membered hetero An aryl group; optionally wherein the C 1-10 alkyl group, the 3-20 membered cycloalkyl group, the 5-20 membered alicyclic group, the 6-20 membered aryl group, the 5-20 membered heteroaryl group are each independently One or more (eg 1, 2, 3 or 4) ground molybdenum is selected from the group consisting of halogen, hydroxy, amino, cyano, carboxy, nitro, C 1-4 alkyl, halo C 1-4 alkyl, hydroxy C 1-4 alkyl, C 1-4 alkoxy, 3-8 membered cycloalkyl, 5-8 membered heteroheterocyclyl, 6-10 membered aryl, 5-10 membered heteroary
  • the compound of formula IA is such that the ring X is not a pyrrolidine ring, a 2-pyrrolidone ring, a morpholine ring, a 3-morpholinone ring, a tetrahydrofuran ring, 2-oxazolidinone ring, tetrahydrothiophene ring and thiophene sulfone ring.
  • a and B together with the attached atom form an aromatic ring (eg, a 6-20 membered aromatic ring, a 6-14 membered aromatic ring, or a 6-10 membered aromatic ring).
  • the aromatic ring is unsubstituted or each independently one or more (eg 1, 2, 3 or 4) selected from halogen (eg fluorine, chlorine, bromine or iodine), hydroxyl, amino, cyanide
  • halogen eg fluorine, chlorine, bromine or iodine
  • a and B are taken together with the attached atom.
  • a benzene ring is formed wherein the benzene ring is unsubstituted or each independently substituted with one or more (eg 1, 2, 3 or 4) substituents selected from halogen (eg, fluorine).
  • R 1 , R 2 , R 3 and R 4 are each independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, cyano, carboxy, nitro, C. 1-10 alkyl and halogenated C 1-10 alkyl.
  • R 1 , R 2 , R 3 and R 4 are each independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, cyano, C 1-6 alkyl. (e.g., methyl) and halo C 1-6 alkyl (e.g., trifluoromethyl).
  • R 1 , R 2 and R 3 are a hydrogen atom.
  • R 4 in the compound of Formula IA is a halogen.
  • the compound of formula IA, R 4 is fluoro.
  • R 5 is selected from the group consisting of hydrogen, oxy, halogen, C 1-10 alkyl, 3-20 membered cycloalkyl, 3-20 membered alicyclic Base, 5-10 membered heteroaryl, RO-, RC(O)-, ROC(O)-, RR'N-, RR'NC(O)-, RS(O) a -, RR'NSO 2 -
  • said C 1-10 alkyl group, 3-20 membered cycloalkyl group, 3-20 membered heteroheterocyclyl group, 5-10 membered heteroaryl group are each independently one or more (eg, 1 , 2, 3 or 4) substituents selected from the group consisting of halogen (preferably fluorine), hydroxyl, amino, cyano, C 1-6 alkyl, C 1-6 alkoxy and 3-8 membered cycloalkyl Where a is 0, 1 or
  • R and R' are each independently selected from the group consisting of hydrogen, C 1-6 alkyl and 3-8 membered cycloalkyl, optionally wherein the C 1-6 alkyl group and the 3-8 membered cycloalkyl group are each Independently one or more (eg 1, 2, 3 or 4) selected from the group consisting of halogen, hydroxy, amino, cyano, C 1-4 alkyl, C 1-4 alkoxy and C 1-4 alkyl The substituent of the sulfonyl group is substituted.
  • R 5 is selected from the group consisting of hydrogen, halogen, C 1-6 alkyl, 3-12 membered cycloalkyl, RC(O)-, and RR'NC ( O)-, optionally, wherein the C 1-6 alkyl group and the 3-12 membered cycloalkyl group are each independently selected from one or more (eg 1, 2, 3 or 4) halogens (preferably fluoro), hydroxy, amino, cyano, C 1-4 alkyl, C 1-4 alkoxy, and 3-8 membered cycloalkyl substituents;
  • R and R' are each independently selected from the group consisting of hydrogen, C 1-6 alkyl and 3-8 membered cycloalkyl.
  • R 5 is selected from the group consisting of hydrogen, halogen, C 1-4 alkyl, 3-6 membered cycloalkyl, RC(O)-, and RR'NC ( O)-, optionally, wherein the C 1-4 alkyl groups are each independently substituted by one or more (eg 1, 2, 3 or 4) substituents selected from halogen (preferably fluorine) and hydroxyl groups ;
  • R and R' are each independently selected from the group consisting of hydrogen, C 1-4 alkyl and 3-6 membered cycloalkyl.
  • the compound of formula IA, R 5 is selected from hydrogen, methyl, trifluoromethyl, cyclopropylmethyl, 2-hydroxy-2-propyl, cyclopropyl, Cyclopropionyl, aminoacyl, carbamoyl and dimethylyl.
  • the compound of formula IA, R 6 is absent or selected from hydrogen and halo C 1-10 alkyl.
  • the compound of formula IA, R 6 is absent or is hydrogen.
  • the compound of formula IA, R 6 is absent.
  • the compound of formula IA, R 6 is hydrogen.
  • Ring X is a five-membered heteroaryl ring and the hetero atom is a nitrogen, or a six-membered nitrogen-containing aliphatic heterocycle; for example, a pyrazole ring or a piperidine ring.
  • the compound of formula IA is Selected from the following structure:
  • the polycyclic ring system of the compound of Formula I-A is selected from the group consisting of:
  • the polycyclic system can have two chiral centers (chiral atoms are indicated by the * symbol), and those skilled in the art will appreciate that the compounds of the present application encompass all of their configurations, such as (R, R), (R, S), (S, S) or (S, R) configuration.
  • the compound has the structure of Formula I-B,
  • a and B together with the attached atoms form an alicyclic, heteroalicyclic, aromatic or heteroaryl ring; optionally wherein the alicyclic, heteroalicyclic, aromatic or heteroaryl ring is independently one or more One (for example 1, 2, 3 or 4) selected from the group consisting of halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aromatic Base, heteroaryl, RO-, ROC(O)-, RC(O)O-, RC(O)-, RR'N-, RC(O)N(R")-, RR'NC(O) - substituted with a substituent of RS(O) a - wherein a is 0, 1 or 2;
  • Ring X is an alicyclic, alicyclic, aromatic or heteroaryl ring; preferably, said ring X is a nitrogen-containing aliphatic heterocyclic or heteroaryl ring; preferably, said ring X is a five- or six-membered nitrogen An aliphatic heterocyclic or heteroaromatic ring; more preferably, the ring X is a five- or six-membered nitrogen-containing heteroaryl ring; for example, a pyrrole ring, an imidazole ring, a pyrazole ring, a triazole ring or a pyridone ring;
  • R 1 , R 2 , R 3 and R 4 are each independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, alkenyl, alkynyl, cycloalkyl, heteroheterocyclyl, Aryl, heteroaryl, RO-, ROC(O)-, RC(O)O-, RC(O)-, RR'N-, RS(O) a -, RC(O)N(R") And RR 'NC(O)-; optionally wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heteroheterocyclyl, aryl, heteroaryl are each independently one or more ( For example, 1, 2, 3 or 4) are selected from the group consisting of halogen, hydroxy, amino, cyano, carboxy, nitro, C 1-4 alkyl, C 1-4 alkoxy, 3-8 membered cycloalkyl, 5
  • R 5 and R 6 are each independently selected from the group consisting of hydrogen, oxy, halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, alkenyl, alkynyl, cycloalkyl, heteroheterocyclyl, Aryl, heteroaryl, RO-, RC(O)-, ROC(O)-, RC(O)O-, RR'N-, RC(O)N(R")-, RR'NC(O )-, ROC(O)N(R')-, RR'NC(O)N(R")-, RS(O) a -, RR'NSO 2 - and RS(O) 2 N(R')
  • the alkyl, alkenyl, alkynyl, cycloalkyl, heteroheterocyclyl, aryl or heteroaryl group are each independently one or more (eg 1, 2, 3 or 4) ) substituents selected from halo, hydroxy,
  • R 5 or R 6 and forming a polycyclic ring system on the ring X to which it is attached, wherein said R 5 or R 6 is selected from the group consisting of a 3-8 membered alicyclic ring, a 5-8 membered aliphatic heterocyclic ring, and a 6-10 membered aromatic group. Ring and 5-10 membered heteroaryl ring;
  • R, R' and R" are each independently selected from the group consisting of hydrogen, C 1-10 alkyl, 3-20 membered cycloalkyl, 5-20 membered heteroheterocyclyl, 6-20 membered aryl and 5-20 membered hetero An aryl group; optionally wherein the C 1-10 alkyl group, the 3-20 membered cycloalkyl group, the 5-20 membered alicyclic group, the 6-20 membered aryl group or the 5-20 membered heteroaryl group are each independently One or more (eg 1, 2, 3 or 4) ground molybdenum is selected from the group consisting of halogen, hydroxy, amino, cyano, carboxy, nitro, C 1-4 alkyl, halo C 1-4 alkyl, hydroxy C 1-4 alkyl, C 1-4 alkoxy, 3-8 membered cycloalkyl, 5-8 membered heteroheterocyclyl, 6-10 membered aryl, 5-10 membered heteroary
  • the limiting condition of the compound of Formula I-B is that
  • R 6 is absent, R 5 is not H, hydroxy, methyl, isopropyl, cyclopropyl, trifluoromethyl, difluoromethyl and trifluoroethyl;
  • the ring X is not a pyrrolidine ring, a 2-pyrrolidone ring, a morpholine ring, a 3-morpholinone ring or a 2-oxazolidinone ring;
  • the compound of the formula IB is not: 4- ⁇ [3-(3,4-dihydro-1H-pyrrolo[1,2-a]pyrazine-2-carbonyl)-4-fluoro-phenyl] Methyl ⁇ -2H-phthalazin-1-one, 4- ⁇ [3-(6,8-dihydro-5H-[1,2,4]triazolo[1,5-a]pyrazine-7 -carbonyl)-4-fluoro-phenyl]methyl ⁇ -2H-phthalazin-1-one, 4- ⁇ [4-fluoro-3-(2-trifluoromethyl-6,8-dihydro-5H) -[1,2,4]triazolo[1,5-a]pyrazine-7-carbonyl)-phenyl]methyl ⁇ -2H-pyridazin-1-one, and 4-[4-fluoro- 3-(5,6-Dihydro-8H-imidazo[1,5-a]pyrazine-7-carbonyl)benzyl]-2H
  • a and B together with the attached atom form an aromatic ring (eg, a 6-20 membered aromatic ring, a 6-14 membered aromatic ring, or a 6-10 membered aromatic ring).
  • the aromatic ring is unsubstituted or each independently one or more (eg 1, 2, 3 or 4) selected from halogen (eg fluorine, chlorine, bromine or iodine), hydroxyl, amino, cyanide
  • halogen eg fluorine, chlorine, bromine or iodine
  • a and B together with the attached atoms form a phenyl ring, wherein the phenyl ring is unsubstituted or independently one or more (eg, 1 , 2, 3 or 4) substituents selected from halogen (e.g., fluorine).
  • halogen e.g., fluorine
  • R 1 , R 2 , R 3 and R 4 are each independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, cyano, carboxy, nitro, C. 1-10 alkyl and halogenated C 1-10 alkyl.
  • R 1 , R 2 , R 3 and R 4 are each independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, cyano, C 1-6 alkyl. (e.g., methyl) and halo C 1-6 alkyl (e.g., trifluoromethyl).
  • R 1 , R 2 and R 3 are a hydrogen atom.
  • R 4 in the compound of formula IB is a halogen.
  • R 4 in the compound of formula IB is fluoro
  • R 5 in the compound of Formula IB is selected from the group consisting of hydrogen, oxy, halogen, C 1-10 alkyl, 3-20 membered cycloalkyl, 3-20 membered alicyclic Base, 5-10 membered heteroaryl, RO-, RC(O)-, ROC(O)-, RR'N-, RR'NC(O)-, RS(O) a -, RR'NSO 2 -
  • said C 1-10 alkyl group, 3-20 membered cycloalkyl group, 3-20 membered heteroheterocyclyl group, 5-10 membered heteroaryl group are each independently one or more (eg, 1 , 2, 3 or 4) substituents selected from the group consisting of halogen (preferably fluorine), hydroxyl, amino, cyano, C 1-6 alkyl, C 1-6 alkoxy and 3-8 membered cycloalkyl Where a is 0, 1 or 2
  • R and R' are each independently selected from the group consisting of hydrogen, C 1-6 alkyl and 3-8 membered cycloalkyl, optionally wherein the C 1-6 alkyl group and the 3-8 membered cycloalkyl group are each Independently one or more (eg 1, 2, 3 or 4) selected from the group consisting of halogen, hydroxy, amino, cyano, C 1-4 alkyl, C 1-4 alkoxy and C 1-4 alkyl The substituent of the sulfonyl group is substituted.
  • R 5 in the compound of Formula IB is selected from the group consisting of hydrogen, oxy, halogen, C 1-6 alkyl, 3-12 membered cycloalkyl, 3-12 alicyclic heterocyclic ring a group, a 5-6 membered heteroaryl group, RO-, RC(O)-, ROC(O)-, RR'N-, RR'NC(O)-, and RS(O) a- , optionally, wherein
  • the C 1-6 alkyl group, the 3-12 membered cycloalkyl group, the 3-12 membered aliphatic heterocyclic group, and the 5-6 membered heteroaryl group are each independently one or more (for example, 1, 2, 3 or 4) a) selected from halogen (preferably fluoro), hydroxy, amino, cyano, C 1-4 alkyl, C 1-4 alkoxy, and 3-8 membered cycloalkyl substituents, wherein a is 0, 1
  • R and R' are each independently selected from the group consisting of hydrogen, C 1-4 alkyl and 3-6 membered cycloalkyl, optionally wherein the C 1-4 alkyl group and the 3-6 membered cycloalkyl group are each Independently one or more (eg 1, 2, 3 or 4) selected from the group consisting of halogen, hydroxy, amino, cyano, C 1-4 alkyl, C 1-4 alkoxy and C 1-4 alkyl The substituent of the sulfonyl group is substituted.
  • R 5 is selected from the group consisting of hydrogen, oxy, halo, C 1-4 alkyl, 3-6 membered cycloalkyl, 3-6 alicyclic heterocycle.
  • the C 1-4 alkyl group, the 3-6 membered cycloalkyl group, the 3-6 membered aliphatic heterocyclic group, and the 5-6 membered heteroaryl group are each independently one or more (for example, 1, 2, 3 or 4) a substituent substituted from a halogen (preferably fluorine), a hydroxyl group, an amino group and a 3-6 membered cycloalkyl group, wherein a is 0, 1 or 2;
  • R and R' are each independently selected from the group consisting of hydrogen, C 1-4 alkyl and 3-6 membered cycloalkyl, optionally wherein the C 1-4 alkyl group and the 3-6 membered cycloalkyl group are each Independently one or more (eg 1, 2, 3 or 4) selected from the group consisting of halogen, hydroxy, amino, cyano, C 1-4 alkyl, C 1-4 alkoxy and C 1-4 alkyl The substituent of the sulfonyl group is substituted.
  • the compound of formula IB in R 5 is selected from hydrogen, bromo, hydroxymethyl, 2-hydroxy-2-propyl, 1-hydroxyethyl, difluoromethyl, Trifluoromethyl, 1,1-difluoroethyl, pentafluoroethyl, 1-aminoethyl, cyclopropylmethyl, cyclopropyldifluoromethyl, cyclopropyl, 1-methyl-1H- Pyrazol-4-yl, 1-methyl-1H-pyrazol-3-yl, 3-methyl-[1,2,4]-oxadiazol-5-yl, 5-methyl-[1, 3,4]-oxadiazol-2-yl, methoxy, cyclopropyloxy, ethoxycarbonyl, cyclopropylamino, aminoacyl, carbamoyl, acetylyl, N,N-dimethylamine An acyl group, a 2-hydroxyethylamino group,
  • the compound of Formula IB R 6 is absent or selected from hydrogen and halo C 1-10 alkyl.
  • the compound of Formula IB R 6 is absent or selected from hydrogen and halo-C 1-6 alkyl group.
  • the compound of Formula IB R 6 is absent.
  • the compound of formula IB in R 6 is selected from hydrogen and halo C 1-4 alkyl (e.g. trifluoromethyl).
  • Ring X in Formula IB is a five-membered heteroaryl ring and the hetero atom is a nitrogen, or a six-membered nitrogen-containing aliphatic heterocycle, preferably, Selected from the following structure:
  • the hydrogen of any of the first aspects of the present application is hydrazine (H) or hydrazine (D).
  • the polycyclic system is selected from the group consisting of:
  • the second aspect of the present invention provides the compound of any one of the first aspect of the present invention, a prodrug, a metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, a hydrate or a solvate thereof.
  • a method of preparing a crystalline form selected from the following methods:
  • R 5 in the formula I is RR'NC(O)-
  • the compound can be synthesized by the following route:
  • Lg in RCO-Lg represents a leaving group such as halogen, -OTs, -OCOR, etc., and the remaining atoms and substituents are as defined in the first aspect of the present application.
  • the condensation reaction operates as follows: 0.8-1.2 equivalents of Compound A and 1.0 equivalent of Compound B, Compound B' or Compound B" (free amine or salt, such as hydrochloric acid The salt) is dissolved in a solvent (for example, DMF or THF), and 0.8-1.2 equivalents of a condensing agent (for example, HATU, EDCI, T3P, etc.) and 2.0-5.0 equivalents of a base (for example, DIPEA, Et 3 N, pyridine, etc.) are added at room temperature.
  • a solvent for example, DMF or THF
  • a condensing agent for example, HATU, EDCI, T3P, etc.
  • a base for example, DIPEA, Et 3 N, pyridine, etc.
  • Compound B is commercially available or can be prepared by routine experimentation in the art.
  • the synthesis of Compound B includes, but is not limited to, the following methods:
  • Lg is a leaving group of a nucleophilic substitution reaction, such as halogen, -OTs, -OCOR, etc.
  • P is a common amino protecting group, such as Boc, Fmoc, Cbz, Alloc, Pht, Tos, Dmb, Bn, etc.
  • R 5 ' is a step may be an addition / reduction reaction of the present application of the R 5 substituent group, such as an ester group, an acyl group and the like, the addition / reduction reagent is a Grignard reagent, for example, NaBH 4, NaBH 3 CN and the like, the remaining atoms or substituents are as defined in the first aspect of the present application.
  • the deprotection of the amino group can be carried out by methods well known in the art, and the application is not limited thereto.
  • the Compound B can be synthesized by the following method:
  • the intermediate 1 and a base are dissolved in a solvent (for example, dichloromethane, tetrahydrofuran, DMF, etc.), and a solution of an acid chloride is added dropwise at a low temperature or a room temperature (the solvent may be dichloromethane, After completion of the reaction, tetrahydrofuran, acetonitrile, etc., extraction, liquid separation, and purification are carried out to obtain intermediates 2-1 and 2-2, and the obtained intermediate is removed. Obtaining the target intermediate after protecting the base;
  • a solvent for example, dichloromethane, tetrahydrofuran, DMF, etc.
  • the intermediate 1 is dissolved in a solvent (e.g., diethyl ether, tetrahydrofuran, etc.), and then a base (e.g., NaH, t-BuOK, LDA, LiHMDS, etc.) is added at a low temperature or room temperature (-80 ° C - room temperature) and reacted at this temperature 10
  • a solution of a halogenated hydrocarbon e.g., diethyl ether, tetrahydrofuran, etc.
  • extraction, drying and purification can obtain intermediates 3-1 and 3-2, and the obtained intermediate is deprotected to obtain the target intermediate;
  • the intermediate 1 is dissolved in a solvent (e.g., toluene, dioxane, DMF, etc.) and then a base (e.g., sodium carbonate, potassium phosphate, potassium carbonate, etc.) is added, and the resulting mixture is added to a palladium catalyst (e.g., Pd 2 (under Pd 2 (under a inert gas). Dba) 3 , Pd(OAc) 2 , Pd(dppf)Cl 2 , etc.) and ligands (eg dppf, xantphos, x-phos, etc.), the reaction can be carried out at room temperature or under heating (60-150 ° C) It can be heated in a microwave reactor. After the reaction is completed, the intermediates 4-1 and 4-2 can be obtained by liquid separation, extraction and purification, and the target intermediate can be obtained by removing the protecting group from the obtained intermediate.
  • a solvent e.g., toluene, diox
  • the SM-1 is dissolved in a solvent (e.g., carbon tetrachloride, chloroform, dichloromethane, etc.) and then a bromine reagent (e.g., bromine, NBS, etc.) is added at room temperature or at a low temperature to obtain the brominated compound Int-1.
  • a bromine reagent e.g., bromine, NBS, etc.
  • This compound is reacted with 2-aminopyrazine in a solvent (e.g., THF, methanol, ethanol, etc.) by adding a base (e.g., potassium carbonate, triethylamine, etc.) at 60-120 °C.
  • a base e.g., potassium carbonate, triethylamine, etc.
  • the intermediate is dissolved in a solvent (for example, methanol, ethanol, isopropanol, etc.) to catalyze hydrogenation to obtain a target product having an ester group, and the reaction may also be added with an acid (for example, acetic acid, hydrochloric acid, etc.) to accelerate or accelerate the reaction;
  • a solvent for example, methanol, ethanol, isopropanol, etc.
  • an acid for example, acetic acid, hydrochloric acid, etc.
  • the compound Int-2 and the amine are dissolved in a solvent (for example, THF, methanol, ethanol, DMF, DMSO, etc.) at room temperature or elevated temperature (60-150 ° C) with a base (such as triethylamine, DIPEA, etc.) or without alkali
  • a base such as triethylamine, DIPEA, etc.
  • the compound Int-3 can be obtained by purification.
  • the reaction can also be carried out in a solvent-free manner or in a microwave reactor, and Int-3 is carried out in a solvent (for example, methanol, ethanol, isopropanol, etc.).
  • Catalytic hydrogenation can obtain an amide target intermediate, and the reaction can also add an acid (such as acetic acid, hydrochloric acid, etc.) to accelerate or promote the reaction;
  • the starting material pyrazine-2-indole and base (such as triethylamine, DIPEA, etc.) are dissolved in a solvent (such as dichloromethane, THF, acetonitrile, etc.) and then the oxalyl chloride monoester is added at low temperature or room temperature, and the reaction is completed.
  • a solvent such as dichloromethane, THF, acetonitrile, etc.
  • the oxalyl chloride monoester is added at low temperature or room temperature, and the reaction is completed.
  • the compound is dissolved in a solvent (methanol, ethanol, DMF, etc.) under heating (40-150 ° C), alkali can be added (such as sodium methoxide, sodium ethoxide, uncle Sodium butoxide, etc.) or no base.
  • reaction After the reaction is completed, it is post-treated and purified to obtain a product of ring closure.
  • the product is subjected to catalytic hydrogenation in a solvent (for example, methanol, ethanol, isopropanol, etc.) to obtain a target product having an ester group, and the reaction may also be added with an acid. (eg acetic acid, hydrochloric acid, etc.) to accelerate or promote the reaction;
  • a solvent for example, methanol, ethanol, isopropanol, etc.
  • an acid eg acetic acid, hydrochloric acid, etc.
  • Int-5 and various amines are dissolved in a solvent (such as THF, methanol, ethanol, DMF, DMSO, etc.) at room temperature or elevated temperature (60-150 ° C) for amine hydrolysis, can add alkali (such as triethylamine, DIPEA, etc. ) or without adding a base, after purification, Int-6 can be obtained.
  • a solvent such as THF, methanol, ethanol, DMF, DMSO, etc.
  • alkali such as triethylamine, DIPEA, etc.
  • the reaction can also be carried out in a solvent-free manner or in a microwave reactor.
  • Int-6 can be catalytically hydrogenated in a solvent (for example, methanol, ethanol, isopropanol, etc.) to obtain a target intermediate of a class of amides.
  • the reaction may also be carried out by adding an acid such as acetic acid, hydrochloric acid or the like to accelerate or promote the
  • the N-protected 3-piperidone is dissolved in a solvent (for example, THF, diethyl ether, etc.) and then added at a low temperature (-90-0 ° C) or at room temperature (for example, KHMDS, LiHMDS, LDA, etc.) and reacted at this temperature 30
  • a solvent for example, THF, diethyl ether, etc.
  • the ester is added, and after the end of the reaction, it is post-treated and purified to obtain a 1,3-diketone derivative, and the derivative is dissolved in a solvent (for example,
  • the reaction of DCM, THF, ethanol, etc. with an aqueous solution of hydrazine, an alcohol solution or a hydrochloride salt can be carried out to obtain an intermediate pyrazole derivative, and the intermediate intermediate can be obtained by removing the protecting group.
  • compound B can be synthesized by the following method:
  • the starting material is dissolved in a solvent (for example, toluene, THF, etc.) and then added with a thio reagent (for example, Lawson's reagent, P 2 S 5 , etc.) at room temperature or elevated temperature, and after completion, the carbonyl group is obtained by thiolation.
  • a solvent for example, toluene, THF, etc.
  • a thio reagent for example, Lawson's reagent, P 2 S 5 , etc.
  • a suitable solvent eg methanol, ethanol, etc.
  • a base eg triethylamine, DIPEA, sodium methoxide, etc.
  • the reaction may also be added with an equivalent amount of mercury acetate, mercury chloride mercury salt or silver carbonate silver salt to accelerate or accelerate the reaction.
  • the ring can be obtained.
  • the azole intermediate can be obtained by removing the protecting group.
  • the starting material pyrazine-2-indole and substituted difluoroacetic acid (SM-2) are dissolved in a strong acid (for example, polyphosphoric acid, etc.) and then combined under heating (for example, 90-150 ° C), after completion of the reaction.
  • a strong acid for example, polyphosphoric acid, etc.
  • the product is dissolved in a solvent (for example, methanol, ethanol, isopropanol, etc.) using a catalyst (for example, palladium carbon, palladium hydroxide carbon, etc.) to carry out catalytic hydrogenation in a hydrogen atmosphere to obtain a target.
  • a catalyst for example, palladium carbon, palladium hydroxide carbon, etc.
  • the reaction may also be added with an acid (such as acetic acid, hydrochloric acid, etc.) to accelerate or promote the reaction;
  • the hydrazide intermediate INT-3 can be obtained by condensation of the starting material pyrazine-2-indole and substituted difluoroacetic anhydride (SM-3) in a solvent such as dichloromethane, tetrahydrofuran or the like.
  • SM-3 substituted difluoroacetic anhydride
  • the product is dissolved in a strong acid (e.g., polyphosphoric acid, etc.) and then combined under heating (e.g., 90-150 ° C), and after completion of the reaction, it is subjected to post-treatment and purification to give the intermediate INT-1.
  • a strong acid e.g., polyphosphoric acid, etc.
  • the application provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound described herein, a prodrug thereof, a metabolite form, a pharmaceutically acceptable salt or ester, or the aforementioned isomer, hydrate,
  • the solvate or crystalline form optionally, further comprises a pharmaceutically acceptable carrier or excipient.
  • the compound, prodrug, metabolite form, pharmacy may also be combined with one or more drugs.
  • the pharmaceutical composition further comprises one or more drugs.
  • the drug is an anti-tumor drug.
  • the anti-tumor drug is selected from the group consisting of temozolomide, doxorubicin, paclitaxel, cisplatin, carboplatin, dacarbazine, topotecan, irinotecan, gemcitabine, bevacizumab , anti-CTLA-4 mAb Ipilimumab, anti-PD-1 mAb pembrolizumab and Nivolumab, and anti-PD-L1 mAb atezolizumab.
  • the carrier includes, but is not limited to, alumina, aluminum stearate, lecithin, serum proteins such as human serum albumin, phosphate, glycerin, sorbic acid, potassium sorbate, saturated Partial glyceride mixture of plant fatty acids, water, protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salt, colloidal silica, magnesium trisilicate, polyvinylpyrrolidone, cellulosic material, poly Ethylene glycol, sodium carboxymethyl cellulose, polyacrylate, beeswax, lanolin.
  • the excipient refers to an addenda other than the main drug in the pharmaceutical preparation.
  • the compound of the present application a prodrug thereof, a metabolite form, a pharmaceutically acceptable salt or ester, or the aforementioned isomer, hydrate, solvate or crystalline form, or a pharmaceutical composition thereof can be given by Drugs: parenteral, topical, intravenous, oral, subcutaneous, intraarterial, intradermal, transdermal, rectal, intracranial, intraperitoneal, intranasal, intramuscular routes or as an inhaler.
  • the compound of the present application may be administered according to the route of administration Formulated into a variety of suitable dosage forms.
  • the compounds of the present application can be formulated into any orally acceptable formulation including, but not limited to, tablets, capsules, aqueous solutions or aqueous suspensions.
  • the carrier used for the tablet generally includes lactose and corn starch, and a lubricant such as magnesium stearate may also be added.
  • the diluent used in the capsule formulation generally comprises lactose and dried cornstarch.
  • Aqueous suspension formulations are usually prepared by admixing the active ingredient with a suitable emulsifier and suspension.
  • some sweeteners, fragrances or colorants may also be added to the above oral formulation forms.
  • the compounds of the present application can be formulated into a suitable ointment, lotion or cream, in which the active ingredient is suspended or dissolved in one or more carriers.
  • Carriers which may be used in ointment preparations include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyethylene oxide, polypropylene oxide, emulsifying wax and water; and detergents or creams which may be used include, but are not limited to, minerals Oil, sorbitan monostearate, Tween 60, cetyl esters wax, hexadecene aryl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • the compounds of the present application can also be administered in the form of a sterile injectable preparation, including sterile injectable aqueous or oily suspension or sterile
  • the injection solution can also be in lyophilized form.
  • carriers and solvents which can be used include water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils may also be employed as a solvent or suspension medium such as a monoglyceride or a diglyceride.
  • the pharmaceutical preparation of the present application includes any preparation pharmaceutically acceptable, such as an oral preparation, a parenteral preparation, and the like.
  • compositions and pharmaceutical preparations of the present application may contain from 0.01 to 2000 mg of the compound of the present application, preferably from 0.1 to 1000 mg of the compound of the present application, preferably from 1 to 800 mg of the compound of the present application, more preferably from 10 to 600 mg of the compound of the present application, particularly preferably 50-500 mg of the compound of the present application.
  • suitable in vitro or in vivo assays are performed to determine the efficacy of the compositions of the present application and whether the administration is suitable for treating a disease or medical condition in a subject. Examples of such assays are described below in connection with specific diseases or medical treatments in non-limiting embodiments.
  • an effective amount of a composition of the present application sufficient to achieve a prophylactic or therapeutic effect is from about 0.001 mg/kg body weight/day to about 10,000 mg/kg body weight/day.
  • the dosage is from about 0.01 mg/kg body weight/day to about 1000 mg/kg body weight/day.
  • the dosage may range from about 0.01 to 1000 mg/kg of host body weight per day, every two days or every three days, more typically from 0.1 to 500 mg/kg of host body weight.
  • An exemplary treatment regimen is once every two days or once a week or once a month.
  • the agent is usually administered multiple times, and the interval between single doses can be daily, weekly, monthly or yearly.
  • the agent can be administered in the form of a sustained release formulation, in which case less dosing frequency is required.
  • the dose and frequency will vary depending on the half-life of the agent in the subject. It can also be different depending on whether it is a preventive treatment or a therapeutic treatment.
  • relatively low doses are administered chronically at relatively low frequency intervals.
  • the application provides the compound, prodrug, metabolite form, pharmaceutically acceptable salt or ester thereof, or the aforementioned isomer, hydrate, solvate or crystalline form, or the present application Use of the pharmaceutical composition in the preparation of a PARP-inhibiting agent.
  • the agent is an agent that inhibits PARP-1.
  • the present application also provides a method of inhibiting PARP activity, the method comprising administering to a cell in need thereof an effective amount of a compound described herein, a prodrug thereof, a metabolite form, pharmaceutically acceptable Salt or ester, or the aforementioned isomer, hydrate, solvate or crystalline form, or a pharmaceutical composition as described herein.
  • the method is for inhibiting PARP-1 activity.
  • the method is for inhibiting PARP-1 activity in a cell.
  • the cell is a cell line or a cell from a subject.
  • the application provides the compound, prodrug, metabolite form, pharmaceutically acceptable salt or ester thereof, or the aforementioned isomer, hydrate, solvate or crystalline form, or the present application
  • the application provides the compound, prodrug, metabolite form, pharmaceutically acceptable salt or ester thereof, or the aforementioned isomer, hydrate, solvate or crystalline form, or the present application
  • the pharmaceutical composition for use in adjuvant treatment of a tumor or for enhancing radiation or chemotherapeutic effects.
  • the application provides a method of adjuvant treatment of a tumor or enhancing the effect of radiation or chemotherapy, the method comprising administering to a subject in need thereof an effective amount of a compound described herein, a prodrug thereof, A metabolite form, a pharmaceutically acceptable salt or ester, or an isomer, hydrate, solvate or crystalline form as described above, or a pharmaceutical composition as described herein.
  • the application provides the compound, prodrug, metabolite form, pharmaceutically acceptable salt or ester thereof, or the aforementioned isomer, hydrate, solvate or crystalline form, or the present application Use of the pharmaceutical composition for the preparation of a medicament for treating a tumor.
  • the application provides the compound, prodrug, metabolite form, pharmaceutically acceptable salt or ester thereof, or the aforementioned isomer, hydrate, solvate or crystalline form, or the present application
  • the pharmaceutical composition for treating a tumor is provided.
  • the application provides a method of treating a tumor, the method comprising providing an effective amount of a compound described herein, a prodrug thereof, a metabolite form, pharmaceutically acceptable to a subject in need thereof Salt or ester, or the aforementioned isomer, hydrate, solvate or crystalline form, or a pharmaceutical composition as described herein.
  • the application provides the compound, prodrug, metabolite form, pharmaceutically acceptable salt or ester thereof, or the aforementioned isomer, hydrate, solvate or crystalline form, or the present application
  • the application provides the compound, prodrug, metabolite form, pharmaceutically acceptable salt or ester thereof, or the aforementioned isomer, hydrate, solvate or crystalline form, or the present application
  • the application provides a method of treating a vascular disease, a neurodegenerative disease, or a nervous system inflammation, the method comprising administering to a subject in need thereof an effective amount of a compound described herein, a precursor thereof A pharmaceutical, metabolite form, pharmaceutically acceptable salt or ester, or an isomer, hydrate, solvate or crystalline form as described above, or a pharmaceutical composition as described herein.
  • the application provides the compound, prodrug, metabolite form, pharmaceutically acceptable salt or ester thereof, or the aforementioned isomer, hydrate, solvate or crystalline form, or the present application
  • the pharmaceutical composition for inhibiting PARP activity in a cell is provided.
  • the cell is a cell line or a cell from a subject.
  • the present application also provides a method of inhibiting PARP activity in a cell, the method comprising administering to the cell an effective amount of a compound described herein, a prodrug thereof, a metabolite form, a pharmaceutically acceptable salt Or an ester, or an isomer, hydrate, solvate or crystalline form as described above, or a pharmaceutical composition as described herein.
  • the method is for inhibiting PARP-1 activity.
  • the cell is a cell line or a cell from a subject.
  • the method is performed in vivo.
  • the method is performed in vitro.
  • the application further provides the compound, a prodrug thereof, a metabolite form, a pharmaceutically acceptable salt or ester, or an isomer, hydrate, solvate or crystalline form, or Use of a pharmaceutical composition as claimed in the preparation of a reagent as a compound, a prodrug thereof, a metabolite form, a pharmaceutically acceptable salt or ester, or the aforementioned isomer, hydrate, solvent
  • the agent or the crystal form assists the inhibition of tumor cell proliferation or the effect of enhancing radiation or chemical inhibition of tumor cell proliferation.
  • the tumor cell is a tumor cell line or a tumor cell from a subject.
  • the agent is for use in an in vivo method.
  • the agent is for use in an in vitro method.
  • the application further provides the compound, a prodrug thereof, a metabolite form, a pharmaceutically acceptable salt or ester, or an isomer, hydrate, solvate or crystalline form, or
  • the pharmaceutical composition of the invention is used for assisting in inhibiting tumor cell proliferation or for enhancing the effect of radiation or chemical inhibition of tumor cell proliferation.
  • the tumor cell is a tumor cell line or a tumor cell from a subject.
  • the compound, a prodrug thereof, a metabolite form, a pharmaceutically acceptable salt or ester, or an isomer, hydrate, solvate or crystalline form, Or the drugs described herein are used in an in vitro method.
  • the present application also provides a method of inhibiting tumor cell proliferation or enhancing the effect of radiation or chemical inhibition of tumor cell proliferation, the method comprising administering to a tumor cell an effective amount of a compound described herein, preceded by a drug, a metabolite form, a pharmaceutically acceptable salt or ester, or an isomer, hydrate, solvate or crystalline form as described above, or a pharmaceutical composition as described herein.
  • the tumor cell is a tumor cell line or a tumor cell from a subject.
  • the method is performed in vitro.
  • the method is performed in vivo.
  • the application further provides the compound, a prodrug thereof, a metabolite form, a pharmaceutically acceptable salt or ester, or an isomer, hydrate, solvate or crystalline form, or Use of the pharmaceutical composition of the invention for the preparation of a reagent for inhibiting tumor cell proliferation.
  • the agent is for use in an in vivo method.
  • the agent is for use in an in vitro method.
  • the application further provides the compound, a prodrug thereof, a metabolite form, a pharmaceutically acceptable salt or ester, or an isomer, hydrate, solvate or crystalline form, or
  • the pharmaceutical composition of the invention is for use in inhibiting tumor cell proliferation.
  • the tumor cell is a tumor cell line or a tumor cell from a subject.
  • the compound, a prodrug thereof, a metabolite form, a pharmaceutically acceptable salt or ester, or an isomer, hydrate, solvate or crystalline form, Or the medicine described in this application The composition is used in an in vivo method.
  • the present application also provides a method of inhibiting proliferation of a tumor cell, the method comprising administering to the cell an effective amount of a compound described herein, a prodrug thereof, a metabolite form, a pharmaceutically acceptable salt or An ester, or an isomer, hydrate, solvate or crystalline form as described above, or a pharmaceutical composition as described herein.
  • the tumor is selected from the group consisting of breast cancer, ovarian cancer, colorectal cancer, melanoma, lung cancer, gastrointestinal stromal tumor, brain cancer, cervical cancer, pancreatic cancer, prostate cancer, Gastric cancer, chronic myeloid leukemia, liver cancer, lymphoma, peritoneal cancer, soft tissue sarcoma, neuroendocrine tumors, and glioblastoma.
  • the tumor cell is selected from the group consisting of a breast cancer cell, an ovarian cancer cell, a colorectal cancer cell, a melanoma cell, a lung cancer cell, a gastrointestinal stromal tumor cell, a brain cancer cell, a cervix Cancer cells, pancreatic cancer cells, prostate cancer cells, gastric cancer cells, chronic myeloid leukocytes, liver cancer cells, lymphoma cells, peritoneal cancer cells, soft tissue sarcoma cells, neuroendocrine tumor cells, and glioblastoma cells.
  • the tumor cell is a tumor cell line or a tumor cell from a subject.
  • Tumors described herein include malignant and benign tumors, and correspondingly, the tumor cells include malignant and benign tumor cells.
  • the subject described herein is a mammal, such as a bovine, equine, ovine, porcine, canine, feline, rodent, primate; wherein, particularly preferred The subject is a human.
  • hydrogen as used herein and hydrogen in each of the groups includes hydrazine (H), hydrazine (D), hydrazine (T).
  • alkyl refers to a straight or branched saturated hydrocarbon group, such as C 1-10 alkyl, C 1-6 alkyl or C 1-4 alkyl, and non-limiting examples include methyl, ethyl , propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, hexyl and the like.
  • the alkyl group is unsubstituted or further substituted with a substituent to form a substituted alkyl group, and the substituent may be selected from a halogen, a hydroxyl group, an alkoxy group, a cycloalkyl group, an aliphatic heterocyclic group, an aryl group or a heteroaryl group.
  • Non-limiting examples of substituted alkyl groups include halo C 1-4 alkyl, hydroxymethyl, 2-hydroxyethyl, 2-hydroxypropan-2-yl, 3-hydroxypropyl, methoxymethyl, 2 -methoxyethyl, 3-methoxypropyl, ethoxymethyl, 2-ethoxyethyl, 3-ethoxypropyl, cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, cyclohexyl Methyl, 2-cyclopropylethyl, 2-cyclobutylethyl, 2-cyclopentylethyl, 2-cyclohexylethyl, 3-cyclopropylpropyl, 3-cyclobutylpropyl, 3-cyclopentylpropyl, 3-cyclohexylpropyl, benzyl, pyridin-2-methyl and the like.
  • alkenyl refers to a straight or branched chain hydrocarbon radical containing at least one carbon to carbon double bond, such as a C 2-10 alkenyl, C 2-6 alkenyl or C 2-4 alkenyl group, without limitation. Examples include ethenyl, propenyl, butenyl, 2-methylpropenyl, pentenyl, 2-methylbutenyl, 3-methylbutenyl, hexenyl, 2-methylpentenyl , 3-methylpentenyl, 4-methylpentenyl, 2-ethylbutenyl, and the like.
  • alkynyl refers to a straight or branched chain hydrocarbon radical containing at least one carbon-carbon triple bond, such as a C 2-10 alkynyl group, a C 2-6 alkynyl group or a C 2-4 alkynyl group, without limitation. Examples include ethynyl, propynyl, butynyl, pentynyl, 3-methylbutynyl, hexynyl, 3-methylpentynyl and the like.
  • cycloalkyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon group, for example a cycloalkyl group containing from 3 to 20 carbon atoms, for example a cycloalkyl group containing from 3 to 12 carbon atoms.
  • a cycloalkyl group having 3 to 10 carbon atoms for example, a cycloalkyl group having 3 to 8 carbon atoms
  • a cycloalkyl group having 3 to 6 carbon atoms for example, a ring having 5 to 8 carbon atoms alkyl.
  • Non-limiting examples of monocycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatrienyl, cyclooctyl Wait.
  • Polycyclic cycloalkyl groups include spiro, fused, and bridged cycloalkyl groups.
  • aliphatic heterocyclyl refers to a saturated or partially saturated cyclic hydrocarbon radical comprising at least one heteroatom selected from the group consisting of N, O and S.
  • an aliphatic heterocyclic group containing 3-20, 3-12, 3-8, 3-6, 5-20, 5-12, 5-8 or 5-6 ring members.
  • Non-limiting examples of aliphatic heterocyclic groups include pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl and the like.
  • aryl refers to a monocyclic or fused polycyclic (eg bicyclic or tricyclic) group having an aromatic character, preferably a 6-20 membered aryl group, preferably a 6-14 membered aryl group, more preferably 6-.
  • aromatic character preferably a 6-20 membered aryl group, preferably a 6-14 membered aryl group, more preferably 6-.
  • 10-membered aryl, aryl groups include, but are not limited to, phenyl and naphthyl and the like.
  • heteroaryl refers to a 5-20 membered aromatic heterocyclic group substituted with at least one hetero atom selected from N, O or S, preferably 5-10 members, preferably 5-6 members. base.
  • heteroaryl groups include furyl, thienyl, pyrrolyl, imidazolyl, oxazolyl, thiazolyl, triazolyl, tetrazolyl, pyridyl, pyridazinyl, pyrimidinyl and pyridyl Azinyl and the like.
  • alicyclic refers to a saturated or partially saturated carbocyclic ring having no aromatic character. For example, it includes a 3-20-membered alicyclic ring, a 3-12-membered alicyclic ring, a 3-8-membered alicyclic ring, a 4-membered alicyclic ring, a 5-membered alicyclic ring, a 6-membered alicyclic ring, and a 7-membered alicyclic ring.
  • aliphatic heterocycle refers to an aliphatic cyclic group wherein at least one ring member is a hetero atom selected from N, O and S.
  • an aliphatic heterocyclic ring containing one or two N atoms for example, an aliphatic heterocyclic ring containing only one O atom, for example, an aliphatic heterocyclic ring containing only one S atom, or the like.
  • it includes a 3-20-membered alicyclic ring, a 3-12-membered alicyclic ring, a 3-8-membered alicyclic ring, a 4-membered alicyclic ring, a 5-membered alicyclic ring, a 6-membered alicyclic ring, a 7-membered alicyclic ring, and the like. .
  • aromatic ring refers to an aromatic ring in which all ring members are carbon atoms. For example, it includes a 6-20 membered aromatic ring, a 6-14 membered aromatic ring, and a 6-10 membered aromatic ring.
  • heteroaryl ring means that at least one ring member is a hetero atom selected from N, O and S.
  • a fragrant cyclic group for example, a heteroaryl ring containing 1 or 2 N atoms, for example, a heteroaryl ring containing only one O atom, for example, a heteroaryl ring containing only one S atom, or the like.
  • it includes a 5-20 membered heteroaryl ring, a 5-14 membered heteroaryl ring, a 5-10 membered heteroaryl ring, a 5-6 membered heteroaryl ring, and the like.
  • halogen as used herein includes fluoro, chloro, bromo and iodo.
  • RO- examples include methoxy, ethoxy, propoxy, cyclopropoxy, and the like.
  • Non-limiting examples of "ROC(O)-" herein include methoxycarbonyl, ethoxycarbonyl, propoxycarbonyl, isopropoxycarbonyl, t-butoxycarbonyl, and the like.
  • Non-limiting examples of "RC(O)O-" herein include formyloxy, acetoxy, propionyloxy and the like.
  • Non-limiting examples of "RC(O)-" herein include formyl, acetyl, propanoyl, cyclopropanoyl and the like.
  • Non-limiting examples of "RR'N-" herein include methylamino, dimethylamino, ethylamino, diethylamino and the like.
  • Non-limiting examples of "RC(O)N(R")-" herein include formylamino, acetylamino, and propionyl groups, and the like.
  • Non-limiting examples of "RR'NC(O)-" herein include carbamoyl, N,N-dimethylamino, ethoxylyl, 2-hydroxyethylamino, 2-methoxyethylamine An acyl group, a 2-hydroxyisobutyryl group, a 2-methanesulfonylethylamino group or the like.
  • RS(O) a - includes a substituted sulfonyl group, a substituted sulfinyl group, and a substituted fluorenyl group, wherein non-limiting examples of the substituted sulfonyl group include a methylsulfonyl group, an ethylsulfonyl group, a benzenesulfonyl group, P-toluenesulfonyl and the like.
  • substituted sulfinyl groups include methylsulfinyl, ethylsulfinyl, phenylsulfinyl, p-tolylsulfinyl and the like.
  • substituted fluorenyl groups include formazan, phenyl fluorenyl, and the like.
  • RR'NS(O) 2 - means a substituted aminosulfonyl group
  • a non-limiting example of RR'NS(O) 2 - includes a dimethylaminosulfonyl group and the like.
  • RS(O) 2 N(R')- means a substituted sulfonamide group such as an alkylsulfonylamino group, an arylsulfonylamino group or the like.
  • Non-limiting examples of RS(O) 2 N(R')- include methanesulfonylamino, ethanesulfonylamino, benzenesulfonylamino and p-toluenesulfonylamino groups, and the like.
  • oxy in this context
  • the oxy group may be substituted with a carbonyl group on a carbon atom on a group such as a cycloalkyl group, an aliphatic heterocyclic group, an aryl group or a heteroaryl group, or may be present in the enol form, for example, pyrrolidone, pyridin-2-one, hydrazine. Structure such as azine-1-one.
  • isomeric forms of the compounds of formula I herein include all possible isomeric forms of the compounds of formula I herein, such as enantiomers, diastereomers, epimers, cis and trans isomers, Conformational isomers, etc.
  • enantiomers of the R and S configurations, and cis and trans isomers of the Z and E configurations are within the scope of the present application.
  • room temperature as used herein is also referred to as ambient temperature or general temperature and generally refers to room temperature, preferably from 20 ° C to 38 ° C. For example, 20 ° C to 30 ° C, for example, 20 ° C to 25 ° C.
  • the compound of the formula I or a pharmaceutically acceptable salt thereof may also form a solvate such as a hydrate, an alcoholate or the like.
  • the compound of the formula I or a pharmaceutically acceptable salt thereof may also exist in the form of a crystal, wherein the crystal refers to an arrangement in which molecules, atoms or ions constituting the compound are repeated in a regular cycle in a space, and the arrangement thereof has a periodicity of a three-dimensional space. Repeatedly at a certain distance.
  • the compounds may exist in two or more crystalline states, and the molecules having the same structure are crystallized into different solid forms, which are called "polymorphs", that is, polymorphs or polymorphs.
  • crystal form When referring to a particular crystalline form or polymorph, collectively referred to as "crystal form", the term "crystalline form" as used herein, the application encompasses any crystalline form of the compound of Formula I or a pharmaceutically acceptable salt thereof.
  • an effective amount refers to an amount sufficient to achieve the desired prophylactic and/or therapeutic effect, for example, to achieve an amount that prevents or reduces the symptoms associated with the condition to be treated.
  • treating refers to both therapeutic treatment and prophylactic measures, the purpose of which is to prevent or delay (reduce) the disease state or condition to which it is directed. If the subject receives a therapeutic amount of a compound, or an isomer, solvate, pharmaceutically acceptable salt thereof, or pharmaceutical composition thereof, as described herein, the subject has one or more indications and symptoms Subjects exhibit an observable and/or detectable reduction or improvement and the subject is successfully "treated”. It will also be understood that the prevention or treatment of the disease state or condition includes not only complete prevention or treatment, but also failure to achieve complete prevention or treatment, but achieving some biological or medical related results.
  • vascular disease mainly refers to myocardial ischemia/reperfusion injury, various forms of heart failure after injury, cardiomyopathy, circulatory shock, cardiovascular aging, cardiovascular complications of diabetes, cardiac hypertrophy, atherosclerosis Hardening, vascular remodeling, angiogenesis.
  • neurodegenerative disease and inflammation of the nervous system mainly refers to stroke, brain trauma, neurodegenerative diseases (Parkinson's disease, Alzheimer's disease and the like) caused by the harmful effects of oxidation and nitrosation stress.
  • Alzheimer's disease and the like mainly refers to stroke, brain trauma, neurodegenerative diseases (Parkinson's disease, Alzheimer's disease and the like) caused by the harmful effects of oxidation and nitrosation stress.
  • Amyotrophic lateral sclerosis and inflammation of the nervous system such as multiple sclerosis.
  • the compound of the present application has excellent long-acting property, whereby the number of administrations can be reduced, and patient compliance can be improved.
  • Figure 1 is a NOE map of the product of Example 19 of the present application.
  • the structure of the compound is determined by nuclear magnetic resonance ( 1 H NMR) or mass spectrometry (MS).
  • the MS was assayed using an Agilent (ESI) mass spectrometer, manufacturer: Agilent, model: Agilent 6120B.
  • ESI Agilent
  • a high performance liquid phase was prepared using Shimadzu LC-8A preparative liquid chromatograph (YMC, ODS, 250 x 20 mml column).
  • the thin layer chromatography silica gel plate was prepared by using an aluminum plate (20 ⁇ 20 cm) manufactured by Merck, and was purified by thin layer chromatography using a GF254 (0.4-0.5 nm) manufactured by Yantai.
  • reaction was monitored by thin layer chromatography (TLC) or LCMS using the developing solvent system: dichloromethane and methanol system, n-hexane and ethyl acetate system, petroleum ether and ethyl acetate system, solvent volume ratio according to compound
  • TLC thin layer chromatography
  • LCMS LCMS using the developing solvent system: dichloromethane and methanol system, n-hexane and ethyl acetate system, petroleum ether and ethyl acetate system, solvent volume ratio according to compound
  • the polarity is adjusted differently or adjusted by adding triethylamine or the like.
  • the microwave reaction was carried out using a Biotage Initiator + (400 W, RT - 300 ° C) microwave reactor.
  • Column chromatography generally uses Qingdao Ocean 200-300 mesh silica gel as a carrier.
  • the system of the eluent includes: dichloromethane and methanol systems, n-hexane and ethyl acetate systems, and the volume ratio of the solvent is adjusted depending on the polarity of the compound, and may also be adjusted by adding a small amount of triethylamine.
  • the temperature of the reaction is room temperature (20 ° C - 30 ° C).
  • the specific conditions are not specified in the examples, and are carried out according to the conventional conditions or the conditions recommended by the manufacturer.
  • the reagents or instruments used are not indicated by the manufacturer, and are conventional products that can be obtained commercially.
  • the reagents used in the present application were purchased from companies such as Acros Organics, Aldrich Chemical Company, and Tebe Chemical.
  • DIPEA N,N-diisopropylethylamine
  • NOE Nuclear Overhauser Effect, the Overhauser nuclear effect
  • NMP N-methylpyrrolidone
  • HATU 2-(7-azobenzotriazole)-N,N,N',N'-tetramethylurea hexafluorophosphate
  • T3P propylphosphoric anhydride.
  • Example 2 The title compound was synthesized by the method of Example 1, substituting 4,5,6,7-tetrahydro-2H-pyrrolo[4,3-c]pyridine for the 4,5,6,7-four used in Example 1. Hydrogen-2H-pyrazolo[3,4-c]pyridine.
  • Example 2 The synthesis was carried out by the method of Example 1, and the 4,5,6 used in Example 1 was replaced with methyl 5,6,7,8-tetrahydro-imidazo[1,2-a]pyrazine-2-carboxylate. , 7-tetrahydro-2H-pyrazolo[3,4-c]pyridine.
  • the second step the compound N-methyl-7-[2-fluoro-5-(4-oxo-3,4-dihydropyridazin-1-methyl)benzoyl)-5,6-dihydrogen -8H-imidazo[1,2-a]pyrazine-2-carboxamide
  • the first step of the compound 7-[2-fluoro-5-(4-oxo-3,4 dihydro-phthalazin-1-methyl)-benzoyl]-5,6-dihydro-8H- Methyl imidazo[1,2-a]pyrazine-2-carboxylate (100 mg, 0.21 mmol) was dissolved in methylamine in methanol (30%, 5 mL), and the obtained mixture was reacted at 85 ° C overnight. After the reaction was completed by TLC, the reaction mixture was concentrated, and the residue was poured into water and extracted with dichloromethane. The organic phase was dried over anhydrous sodium sulfate.
  • the second step the compound N-methyl-7-[2-fluoro-5-(4-oxo-3,4-dihydro-pyridazine-1-methyl)-benzoyl]-5,6- Dihydro-8H-[1,2,4]triazolo[4,3-a]pyrazine-3-carboxamide
  • Second step 4-[3-(2-cyclopropanoyl-2,4,5,6-tetrahydro-pyrazolo[3,4-c]pyridine-6-carbonyl)-4-fluorobenzyl ]-2H-pyridazin-1-one
  • the third step 4-[3-(2-cyclopropylmethyl-2,4,5,7-tetrahydro-pyrazolo[3,4-c]pyridine-6-carbonyl)-4-fluorobenzyl -2H-phthalazin-1-one
  • the third step 4-[4-fluoro-3-(2-methyl-2,4,5,7-tetrahydro-pyrazolo[3,4-c]pyridine-6-carbonyl)-benzyl] -2H-phthalazin-1-one
  • the nuclear magnetic spectrum confirmed that the sample was a mixture of two isomers in a ratio of 3:1.
  • Example 1 The title compound was synthesized by the method of Example 1 and replaced with ethyl 5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazine-3-carboxylate. 4,5,6,7-tetrahydro-2H-pyrazolo[3,4-c]pyridine used in Example 1.
  • Example 2 The title compound was synthesized by the method of Example 1, substituting 3-trifluoromethyl-4,5,6,7-tetrahydro-1H-pyrazolo[3,4-c]piperidine for the use in Example 1. 4,5,6,7-tetrahydro-2H-pyrazolo[3,4-c]pyridine.
  • Example 1 The title compound was synthesized using the experimental procedure of Example 1 and was carried out by substituting ethyl 2-trifluoromethyl-5,6,7,8-tetrahydro-imidazo[1,2-a]pyrazine-3-carboxylate. 4,5,6,7-tetrahydro-2H-pyrazolo[3,4-c]pyridine as used in Example 1.
  • Example 2 The title compound was synthesized by the method of Example 1, and the 4,5 used in Example 1 was replaced with 4,5,6,7-tetrahydro-pyrrolo[1,5-a]pyrazin-2-yl)-methanol. 6,7-tetrahydro-2H-pyrazolo[3,4-c]pyridine.
  • Example 2 The title compound was synthesized by the method of Example 1, substituting 1,2,3,4-tetrahydro-pyrido[1,2-a]pyrazine-6-one for 4,5,6 as used in Example 1. 7-Tetrahydro-2H-pyrazolo[3,4-c]pyridine.
  • Example 2 The title compound was synthesized by the method of Example 1 and replaced with 3-bromo-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazine in Example 1. 4,5,6,7-tetrahydro-2H-pyrazolo[3,4-c]pyridine used.
  • Step 3 4- ⁇ 4-Fluoro-3-[3-(1-hydroxy-1-methyl-ethyl)-5,6-dihydro-8H-imidazo[1,2-a]pyrazine -7-carbonyl]-benzyl ⁇ -2H-phthalazin-1-one
  • the second step the synthesis was carried out by the method of the second step of Example 4.
  • the second step the synthesis was carried out by the method of the second step of Example 4.
  • Second step 2-(2-trifluoromethyl-5,6,7,8-tetrahydro-imidazo[1,2-a]pyrazin-3-yl)-propyl-2-ol
  • the third step 4- ⁇ 4-fluoro-3-[3-(1-hydroxy-1-methyl-ethyl)-2-trifluoromethyl-5,6-dihydro-8H-imidazo[1 ,2-a]pyrazine-7-carbonyl]-benzyl ⁇ -2H-phthalazin-1-one
  • the third step 4-[4-fluoro-3-(3-methoxy-5,6-dihydro-8H-[1,2,4]triazolo[4,3-a]pyrazine- 7-carbonyl)-benzyl]-2H-phthalazin-1-one
  • the compound obtained in the first step was dissolved in 2-acetyl-5,6-dihydro-8H-[1,2,4]triazolo[4,3-a]pyrazine-7-carbonate tert-butyl ester. Methyl chloride was then added to the trifluoroacetic acid until the reaction was complete. After the reaction mixture was concentrated, the resulting mixture was used directly in the next step.
  • the third step 4-[3-(3-acetyl-5,6-dihydro-8H-[1,2,4]triazolo[4,3-a]pyrazine-7-carbonyl)- 4-fluorobenzyl]-2H-phthalazin-1-one
  • the product obtained in the first step was dissolved in 2 mL of methanol, then a saturated solution of hydrogen chloride in 1,4-dioxane (2 mL, 4N) was added to the reaction at room temperature until the reaction was completed, and the solvent was evaporated under reduced pressure. reaction.
  • the third step 4- ⁇ 4-fluoro-3-[3-(1-methyl-1H-pyrazol-4-yl)-5,6-dihydro-8H-imidazo[1,2-a] Pyrazin-7-carbonyl]-benzyl ⁇ -2H-phthalazin-1-one
  • Example 38 For the synthesis method, see the first step of Example 38 using 3-bromo-5,6-dihydro-8H-[1,2,4]triazolo[4,3-a]pyrazine-7-carbonate tert-butyl ester. The 3-bromo-5,6-dihydro-8H-imidazo[1,2-a]pyrazine-7-carbonate tert-butyl ester in Example 38 was replaced.
  • the third step is 4- ⁇ 4-fluoro-3-[3-(1-methyl-1H-pyrazol-4-yl)-5,6-dihydro-8H-[1,2,4]triazole And [4,3-a]pyrazine-7-carbonyl]-benzyl ⁇ -2H-pyridazin-1-one
  • Example 33 For the synthesis method, the first step of Example 33 was carried out, and sodium methoxide used in the first step of Example 33 was replaced with sodium methanethiolate, and the solvent was changed to DMF.
  • the third step 4-[4-fluoro-3-(3-methylindolyl-5,6-dihydro-8H-[1,2,4]triazolo[4,3-a]pyrazine-7 -carbonyl)-benzyl]-2H-phthalazin-1-one
  • Example 33 For the synthesis method, the first step of Example 33 was carried out, and sodium methoxide used in the first step of Example 33 was replaced with sodium methanethiolate, and the solvent was changed to DMF.
  • the third step 3-methylsulfonyl-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazinetrifluoroacetate
  • Step 4 4-[4-Fluoro-3-(3-methylsulfonyl-5,6-dihydro-8H-[1,2,4]triazolo[4,3-a]pyrazine -7-carbonyl)-benzyl]-2H-phthalazin-1-one
  • the third step 4- ⁇ 4-fluoro-3-[3-(2-hydroxypropyl-2-yl)-5,6-dihydro-8H-[1,2,4]triazolo[4 ,3-a]pyrazine-7-carbonyl]-benzyl ⁇ -2H-phthalazin-1-one
  • 6-tert-Butoxycarbonyl-4,5,6,7-tetrahydro-1H-pyrazolo[3,4-c]pyridine-3-carboxylic acid (267 mg, 1 mmol) was dissolved in 5 mL of methanol and then slowly added 1 mL of thionyl chloride. The resulting solution was refluxed overnight, and after methanol was taken to leave, the methanol was removed and the residue was used directly to the next reaction.
  • Step 5 4- ⁇ 4-Fluoro-3-[3-(2-hydroxypropyl-2-yl)-1,4,5,7-tetrahydro-pyrazolo[3,4-c]pyridine -6-carbonyl]-benzyl ⁇ -2H-phthalazin-1-one
  • Example 29 For the synthesis, see the third step of Example 29, using 2-(4,5,6,7-tetrahydro-1H-pyrazolo[3,4-c]pyridin-3-yl)propyl-2-ol trifluoro Replacement of 2-(5,6,7,8-tetrahydro-imidazo[1,2-a]pyrazin-3-yl)-propyl-2-ol trifluoroacetic acid in Example 29 with acetate salt.
  • the third step 4- ⁇ 4-fluoro-3-[3-(1-hydroxyethyl)-5,6-dihydro-8H-[1,2,4]triazolo[4,3-a Pyrazin-7-carbonyl]-benzyl ⁇ -2H-pyridazin-1-one
  • Example 29 For the synthesis, see the third step of Example 29, using 1-(5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazin-3-yl)ethanol. Substituting trifluoroacetate for 2-(5,6,7,8-tetrahydro-imidazo[1,2-a]pyrazin-3-yl)-propyl-2-ol trifluoro in Example 29. Acetate.
  • Step 2 3-(1-Methyl-1H-pyrazol-3-yl)-5,6-dihydro-8H-[1,2,4]triazolo[4,3-a]pyridin Pyridin-7-tert-butyl carbonate
  • Example 38 For the synthesis, the first step of Example 38 was followed by substituting 1-methyl-1H-pyrazole-3-boronic acid for 1-methyl-1H-pyrazole-4-boronic acid in Example 38.
  • Step 4 4 ⁇ 4-Fluoro-3-[3-(1-methyl-1H-pyrazol-3-yl)-5,6-dihydro-8H-[1,2,4]triazole And [4,3-a]pyridyl Pyrazin-7-carbonyl]-benzyl ⁇ -2H-phthalazin-1-one
  • the third step 4- ⁇ 3-[3-(1,1-difluoroethyl)-5,6-dihydro-8H-[1,2,4]triazolo[4,3-a] Pyrazin-7-carbonyl]-4- Fluorobenzyl ⁇ -2H-pyridazin-1-one (Compound 46)
  • Step 3 4-[4-Fluoro-3-(3-pentafluoroethyl-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazine -7-carbonyl)benzyl]-2H-phthalazin-1-one
  • Enzyme activity screening of PARP-1 inhibitors was performed using the PARP1 chemiluminescence kit from BPS Bioscience.
  • Kit PARP1Chemiluminescent Assay Kit
  • Pretreatment of the plate remove the microplate from the package, then add 50 ⁇ L/well of 1 ⁇ PARP buffer diluted 1 ⁇ histone, incubate overnight at 4°C; discard the liquid in the microplate the next day, and use 200 ⁇ L Wash the plate with PBST (1 ⁇ PBS and 1% Triton X-100) to remove all the cleaning solution; then add 200 ⁇ L of Stop Buffer to each well, incubate for 90 min at room temperature, discard the liquid; and wash the plate with 200 ⁇ L/well PBST. Finally remove all cleaning fluid.
  • Detection After completion of the reaction, 50 ⁇ L of diluted Strep-HRP was added to each well, and cultured at room temperature; the washing solution was removed after washing the plate with PBST; HRP substrates A and B were mixed, and then 100 ⁇ L of each well was added; (Luminometric Measurement) The test compound was tested for PARP1 kinase inhibitory activity.
  • the compounds of the present application have a significant inhibitory effect on PARP1 kinase activity, and The compound inhibiting activity is significantly better than the positive drug.
  • the effect of the compound on cell proliferation was determined by the CCK-8 method.
  • Kit CCK-8 Kit (Cell Counting Kit-8)
  • Cell culture The cells were cultured according to the following conditions, and the cells were counted, counted by a cell counter, and the cells were adjusted to the desired concentration according to the following requirements, and then 100 ⁇ L of the cells were seeded per well, and administered 24 hours after the inoculation.
  • the mother liquor was prepared by dissolving the experimental drug and olaparibil in DMSO, and the appropriate mother liquor was aspirated into the culture solution to be mixed, and the drug solution was configured to the corresponding incubation concentration.
  • Incubation time Incubation for 7 days in the incubator after administration.
  • Detection After the incubation, the fresh medium was replaced with 200 ⁇ L/well in the medium for 2 h, then 20 ⁇ L/well of CCK8 was added for more than 3 h, and the absorbance was measured at a wavelength of 450 nm and a reference wavelength of 650 nm. The IC 50 value was calculated using GraphPad.
  • the compounds in Table 5-1 were superior to olaparib in inhibiting breast cancer MDA-MB-468 tumors.
  • the compounds of the present application essentially have similar inhibitory effects on the breast cancer MDA-MB-468 tumor as the above compounds.
  • the compounds of the present application have similar inhibitory effects on breast cancer MDA-MB-436, MDA-MB-453 tumors.
  • pancreatic cancer Capan-1 tumors The inhibitory effect of the compounds in Table 5-2 on pancreatic cancer Capan-1 tumors was superior to that of olaparib.
  • the compounds of the present application essentially have similar inhibitory effects as the above compounds against pancreatic cancer Capan-1 tumors.
  • the inhibitory effects of the compounds in Table 5-3 on rectal cancer HCT116 tumors were superior to those of olaparib.
  • the compounds of the present application have substantially similar inhibitory effects as the above compounds against rectal cancer HCT116 tumors.
  • the compounds of the present application have excellent inhibitory activity against cancer cells such as breast cancer, colorectal cancer and pancreatic cancer, and exhibit good antitumor effects.
  • LC-MS/MS the column was a Waters X-Bridge C18 column (21 mm*50 mm, 3.5 ⁇ m); the mobile phase A phase was water + 2 mM ammonium acetate, the phase B was methanol + 2 mM ammonium acetate, and the flow rate was 0.4 mL/min.
  • the column temperature is 40 °C.
  • the ion source is used as the ESI source positive ion mode, and the scanning mode is multiple reaction monitoring (MRM).
  • the pharmacokinetic parameters of the compounds of the present application in rats were superior to olaparib according to the results of the IV mode of administration.
  • the pharmacokinetic parameters of the compounds of the present application in rats were superior to those of olaparib.
  • the potassium channel encoded by the human Ether-a-go-go Related Gene mediates a delayed rectifier potassium current (IKr). IKr inhibition is the most important mechanism for drug-induced QT interval prolongation.
  • the criterion is that if the IC50 of the test compound is >30 ⁇ M, it is determined that the test compound has no inhibitory effect on hERG.
  • KM mice were administered by gavage, and the toxicity of a single administration of the test compound was examined to determine the maximum tolerated dose (MTD).
  • MTD maximum tolerated dose
  • the dose was set to Compound 1: 300 mg/kg; Compounds 2, 3, 6: 200, 300 mg/kg, administered by single gavage, the vehicle was 10% DMSO plus 50% PEG, and 7 days after administration.
  • This example was used to evaluate the inhibitory effectiveness of the different routes of administration of the compounds of the present application on tumor proliferation.
  • the human pancreatic cancer cell line Capan-1 subcutaneous xenograft mice were examined for their effects on pancreatic cancer Capan-1 tumor-bearing mice by administering a sample of the compound of the present application by a PO administration route.
  • Tumor-bearing mice with a tumor volume of 100-200 mm 3 were randomly divided into 8 groups, 7 rats/group, and the administration volume was 10 ml/kg, once a day for about two weeks. Tumor volume was measured twice a week after administration.
  • the antitumor effect of the test compound was evaluated by the tumor growth inhibition rate TGI (%).
  • TGI (%) [1- (VT end - VT start) / (VC end - VC start)] * 100%
  • VT is the end of the tumor volume at the end of the treatment group
  • VT start tumor volume mean at the beginning of treatment in the treatment group
  • End of VC mean volume of tumor at the end of the vehicle control experiment
  • VC start mean volume of tumor at the beginning of drug control group administration
  • the compound of the present application has excellent pharmacological effects, and the tumor partially disappears at a dose of 10 ml/kg and the like, and the drug efficacy is superior to the positive control group.
  • composition of the immediate release tablets is shown in Table 10:
  • Standard immediate release tablets are made using direct compression.
  • Compound 1 and lactose, microcrystalline cellulose, croscarmellose sodium and sodium lauryl sulfate were weighed into glass vials to occupy approximately 75% of the volume of the tubules, which were then mixed together for 30 minutes in a tumble mixer.
  • the blended material was sieved through a 40 mesh (425 ⁇ m) sieve and then drum mixed for another 15 minutes.
  • Magnesium stearate was then added and the blend was shaken by hand for about 20 seconds.
  • the resulting mixture was then dispensed into 400 mg aliquots and compressed into tablet cores using a hand press equipped with a 10 mm die with 0.5 ton of target compression.
  • composition of the capsule preparation is shown in Table 11.
  • the preparation method is as follows:
  • the material After re-pulverizing the material of 20 mesh or more, the material is subjected to secondary dry granulation with a material of 60 mesh or less, and the above-mentioned parts by mass of magnesium stearate is added to the granules, and the mixed granules are uniformly mixed to prepare the mixed granules. Capsules are made on the filling machine.

Abstract

本申请涉及酞嗪酮衍生物、其制备方法及用途;具体涉及一种式I化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型,其制备方法及用途,本申请的化合物可以明显提高对肿瘤细胞的增殖抑制作用;可以增加分子体内稳定性以及降低产生毒性代谢物的可能性;或者,可通过降低对酞嗪酮类化合物的结构修饰,降低化合物在体内P450细胞色素酶系作用下的氧化代谢能力,并提高了生物利用率,

Description

酞嗪酮衍生物、其制备方法及用途 技术领域
本申请涉及医药领域,具体涉及酞嗪酮衍生物、其制备方法及用途。
背景技术
聚腺苷二磷酸核糖聚合酶(Poly ADP-Ribose Polymerase,PARP)是一种113kDa的多结构域蛋白质,其通过识别和快速结合断裂的DNA单链或双链参与DNA损伤的信号传导(D’Amours D.et al.,Poly(ADP-ribosyl)ation reactions in the regulation of nuclear functions.Biochem.J.342:249-268(1999))。PARP家族现在包括约18种蛋白质,其中PARP-1(最早发现的成员)和PARP-2是目前发现的仅有的通过发生DNA断裂而刺激其催化活性的酶,这使得它们在该家族中非常独特。而PARP-1是PARP家族中结构最典型,也是研究最多的,其分子量为114KDa,以ADP为底物在受体蛋白(包括自身)上合成聚腺苷酸二磷酸核糖(PRA)(Sakamoto-Hojo E T,Balajee A S.Targeting Poly(ADP)ribose Polymerase I(PARP-I)and PARP-I interacting Proteins for cancer treatment.Anticancer Agents Med Chem,2008,8(4):402-416)。
PARP-1参与DNA损伤修复及转录调节,并被认为是细胞存活和死亡的重要调节因子,还参与肿瘤发生及炎症反应中的一些转录因子的调控(Peralta-Leal A,Rodriguez-Vargas J M,Aguilar-Quesada R,et al.PARP inhibitors:New partners in the therapy of cancer and inflammatory diseases.FreeRadicalBiol Med,2009,47(l):13-26)。目前为止人们已发现PARP-1在多种人类的恶性肿瘤中高表达,如恶性淋巴瘤,乳腺癌,尤文氏肉瘤,肝细胞癌等。
由于PARP-1参与DNA损伤修复,单独应用PARP-1活性抑制剂或与DNA损伤药物联用可以促进癌细胞的死亡。大量的研究已经证实药物抑制或基因敲除PARP-1不仅能避免氧化应激相关疾病引起的组织损伤,还能改善肿瘤病人的预后。单独应用PARP-1抑制剂对DNA损伤修复缺陷的肿瘤(主要是乳腺癌)也有杀伤作用。另外文献还报道了PARP-1抑制剂与血管生成的关系,目前至少有五种PARP抑制剂在体外能抑制血管内皮细胞生长因子(VEGF)诱导的人脐静脉内皮细胞的增殖和迁移以及血管形成。因为大脑很容易受到氧化应激的影响,PARP-1的神经病理学研究也逐渐受到重视。
因此,开发新型PARP抑制剂具有较高的临床应用价值。
发明内容
本申请的发明人通过对酞嗪酮类化合物的构效关系进行系统的研究,发现了一类具有优异PARP抑制活性的化合物,本申请即是基于以上发现而完成。
本申请的第一方面提供式I化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型,
Figure PCTCN2016110007-appb-000001
其中,
A和B与相连的原子一起形成脂环、脂杂环、芳环或杂芳环;任选地,其中所述脂环、脂杂环、芳环或杂芳环各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、羧基、硝基、烷基、卤代烷基、烯基、炔基、环烷基、脂杂环基、芳基、杂芳基、RO-、ROC(O)-、RC(O)O-、RC(O)-、RR’N-、RC(O)N(R”)-、RR’NC(O)-和RS(O)a-的取代基取代,其中a为0、1或2;
D和E各自独立地选自C和N,并与相连的原子一起形成5-10元环X,其中所述环X为脂环、脂杂环、芳环或杂芳环;优选地,所述环X为含氮脂杂环或杂芳环;优选地,所述环X为五元或六元含氮脂杂环或杂芳环;优选地,所述环X为五元或六元含氮杂芳环;所述环X例如为吡咯环、咪唑环、吡唑环、三氮唑环、吡啶酮环或哌啶环;
m和n各自独立地选自0、1、2和3,且m+n=3或4;
R1、R2、R3和R4各自独立地选自氢、卤素、羟基、氨基、氰基、羧基、硝基、烷基、烯基、炔基、环烷基、脂杂环基、芳基、杂芳基、RO-、ROC(O)-、RC(O)O-、RC(O)-、RR’N-、RS(O)a-、RC(O)N(R”)-和RR’NC(O)-;任选地,其中所述烷基、烯基、炔基、环烷基、脂杂环基、芳基或杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、羧基、硝基、C1-4烷基、C1-4烷氧基、3-8元环烷基、5-8元脂杂环基、6-10元芳基、5-10元杂芳基和RSO2-的取代基取代,其中a为0、1或2;
R5和R6各自独立地选自不存在、氢、氧基、卤素、羟基、氨基、氰基、羧基、硝基、烷基、烯基、炔基、环烷基、脂杂环基、芳基、杂芳基、RO-、RC(O)-、ROC(O)-、RC(O)O-、RR’N-、RC(O)N(R”)-、RR’NC(O)-、ROC(O)N(R’)-、RR’NC(O)N(R”)-、RS(O)a-、RR’NSO2-和RS(O)2N(R’)-;任选地,其中所述烷基、烯基、炔基、环烷基、脂杂环基、芳基或杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、羧基、硝基、C1-4烷基、C1-4烷氧基、3-8元环烷基、5-8元脂杂环基、6-10元芳基、5-10元杂芳基和RSO2-的取代基取代,其中a为0、1或2;或者,
R5或R6并在与之相连的环X上形成多环系统,其中,所述R5或R6选自3-8元脂环、 5-8元脂杂环、6-10元芳环和5-10元杂芳环;
R、R’和R”各自独立地选自氢、C1-10烷基、3-20元环烷基、5-20元脂杂环基、6-20元芳基和5-20元杂芳基,任选地,其中所述C1-10烷基、3-20元环烷基、5-20元脂杂环基、6-20元芳基或5-20元杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、羧基、硝基、C1-4烷基、卤代C1-4烷基、羟基C1-4烷基、C1-4烷氧基、3-8元环烷基、5-8元脂杂环基、6-10元芳基、5-10元杂芳基和RSO2-的取代基取代。
在本申请的某些优选实施方案中,所述式I化合物的限制性条件为,
当A和B与相连的碳原子形成未取代的苯环,R1、R2和R3均为H,R4为F时,
1)且当式I中
Figure PCTCN2016110007-appb-000002
Figure PCTCN2016110007-appb-000003
时,R6不存在,R5不为H、羟基、甲基、异丙基、环丙基、三氟甲基、二氟甲基和三氟乙基;
2)且当式I中
Figure PCTCN2016110007-appb-000004
Figure PCTCN2016110007-appb-000005
时,R5和R6不全为H、且当R5或R6其中一个为H时,另一个不为三氟甲基和乙氧羰基;
3)且式I中
Figure PCTCN2016110007-appb-000006
不为
Figure PCTCN2016110007-appb-000007
4)所述环X不为吡咯烷、2-吡咯烷酮、吗啉、3-吗啉酮、四氢呋喃、2-噁唑烷酮、四氢噻吩和噻吩烷砜;并且,
5)所述式I化合物不为:
4-{[3-(3,4-二氢-1H-吡咯并[1,2-a]吡嗪-2-羰基)-4-氟-苯基]甲基}-2H-酞嗪-1-酮;
4-{[3-(6,8-二氢-5H-[1,2,4]三唑并[1,5-a]吡嗪-7-羰基)-4-氟-苯基]甲基}-2H-酞嗪-1-酮;
4-{[4-氟-3-(2-三氟甲基-6,8-二氢-5H-[1,2,4]三唑并[1,5-a]吡嗪-7-羰基)-苯基]甲基}-2H-酞嗪-1-酮;和
4-[4-氟-3-(5,6-二氢-8H-咪唑并[1,5-a]吡嗪-7-羰基)苄基]-2H-酞嗪-1-酮。
在本申请的某些优选实施方案中,式I化合物中所述A和B与相连的原子一起形成芳环(例如6-20元芳环、6-14元芳环或6-10元芳环),其中,所述芳环未被取代或各自独立地被一个或多个(例如1、2、3或4个)选自卤素(例如氟、氯、溴或碘)、羟基、氨基、氰基、羧基、硝基和C1-10烷基的取代基取代。
在本申请的某些优选实施方案中,所述式I化合物中A和B与相连的原子一起形成苯环,其中,所述苯环未被取代或各自独立地被一个或多个(例如1、2、3或4个)选自卤素(例如氟)的取代基取代。
在本申请的某些优选实施方案中,所述式I化合物中R1、R2、R3和R4各自独立地选自氢、卤素、羟基、氨基、氰基、羧基、硝基、C1-10烷基和卤代C1-10烷基。
在本申请的某些优选实施方案中,所述式I化合物中R1、R2、R3和R4各自独立地选自氢、卤素、羟基、氨基、氰基、C1-6烷基(例如甲基)和卤代C1-6烷基(例如三氟甲基)。
在本申请的某些优选实施方案中,所述式I化合物中R1、R2和R3为氢原子。
在本申请的某些优选实施方案中,所述式I化合物中R4为卤素。
在本申请的某些优选实施方案中,所述式I化合物中R4为氟。
在本申请的某些优选实施方案中,所述式I化合物中A和B与相连的原子一起形成苯环,R1、R2和R3为氢,且R4为为氟。
在本申请的某些优选实施方案中,所述式I化合物中R5和R6各自独立地选自不存在、氢、氧基、卤素、C1-10烷基、3-20元环烷基、3-20元脂杂环基、5-10元杂芳基、RO-、RC(O)-、ROC(O)-、RR’N-、RR’NC(O)-、RS(O)a-和RR’NSO2-;任选地,其中所述C1-10烷基、3-20元环烷基、3-20元脂杂环基或5-10元杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素(优选为氟)、羟基、氨基、氰基、C1-6烷基、C1-6烷氧基、3-8元环烷基、3-8元脂杂环基、6-10元芳基和5-10元杂芳基的取代基取代,其中a为0、1或2;
且,
R和R’各自独立地选自氢、C1-6烷基、3-8元环烷基、5-8元脂杂环基、6-10元芳基和5-10元杂芳基;任选地,其中所述C1-6烷基、3-8元环烷基、5-8元脂杂环基、6-10元芳基、5-10元杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、C1-4烷基、卤代C1-4烷基、羟基C1-4烷基、RS(O)2-、C1-4烷氧基、3-8元环烷基、5-8元脂杂环基、6-10元芳基和5-10元杂芳基的取代基取代;或者,R和R’与所连的N原子一起形成4-10元脂杂环基;或者,
R5或R6并在与之相连的环X上形成多环系统,其中,所述R5或R6选自脂杂环(例如5-8元脂杂环)和杂芳环(例如5-10元杂芳环);优选地,所述脂杂环或杂芳环为五元或六元含氮脂杂环或杂芳环;例如为吡咯烷环、吡咯环、咪唑环、吡唑环、三氮唑环、噁唑环、噻唑环、哌啶环、哌嗪环、吡嗪环、吗啉环、硫吗啉环、吡啶环、嘧啶环或哒嗪环。
在本申请的某些优选实施方案中,所述式I化合物中R5和R6各自独立地选自不存在、氢、氧基、卤素(例如F、Cl、Br或I)、C1-6烷基和取代的C1-6烷基(例如甲基、乙基、氟代甲基、二氟甲基、三氟甲基、1,1-二氟乙基、环丙基-二氟甲基、羟甲基、1-羟基乙基、2-羟基乙基、1-氨基乙基、环丙基甲基、2-羟基-2-丙基)、C1-6烷氧基(例如甲氧基、乙氧基、丙氧基、叔丁氧基、环丙基氧基、)、3-8元环烷基(例如环丙基、环丁基、环戊基、环己基)、取代的酰基(例如环丙甲酰基、乙酰基)、氨基和取代的氨基(例如环丙基氨基)、氨酰基和取代的氨酰基(氨酰基、甲氨酰基、N,N-二甲胺酰基、乙氨酰基、2-羟基乙基氨酰基、2-甲氧基乙基氨酰基、2-羟基异丁氨酰基、2-甲磺酰基乙基氨酰基)、取代的氧羰基(例如甲氧羰基、乙氧羰基)、杂芳基和取代的杂芳基(例如吡唑基、1-甲基吡唑基)、脂杂环基和取代的脂杂环基(例如2-甲基-[1.3.4]氧代二唑-5-基、3-甲基-[1.2.4]氧代二唑-5-基)、甲硫基和甲磺酰基。
在本申请的某些优选实施方案中,所述式I化合物中R5选自氢、氧基、卤素、C1-10烷基、3-20元环烷基、3-20元脂杂环基、5-10元杂芳基、RO-、RC(O)-、ROC(O)-、RR’N-、RR’NC(O)-、RS(O)a-和RR’NSO2-;任选地,其中所述C1-10烷基、3-20元环 烷基、3-20元脂杂环基或5-10元杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素(优选为氟)、羟基、氨基、氰基、C1-6烷基、C1-6烷氧基、3-8元环烷基、3-8元脂杂环基、6-10元芳基和5-10元杂芳基的取代基取代,其中a为0、1或2;或者,
R5并在与之相连的环X上形成多环系统,其中,所述R5选自脂杂环(例如5-8元脂杂环)和杂芳环(例如5-10元杂芳环);优选地,所述脂杂环或杂芳环为五元或六元含氮脂杂环或杂芳环;例如为吡咯烷环、吡咯环、咪唑环、吡唑环、三氮唑环、噁唑环、噻唑环、哌啶环、哌嗪环、吡嗪环、吗啉环、硫吗啉环、吡啶环、嘧啶环或哒嗪环;
所述R6不存在或选自氢、氧基、卤素、C1-10烷基、3-20元环烷基、3-20元脂杂环基、5-10元杂芳基、RO-、RC(O)-、ROC(O)-、RR’N-、RR’NC(O)-、RS(O)a-和RR’NSO2-;任选地,其中所述C1-10烷基、3-20元环烷基、3-20元脂杂环基或5-10元杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素(优选为氟)、羟基、氨基、氰基、C1-6烷基、C1-6烷氧基、3-8元环烷基、3-8元脂杂环基、6-10元芳基和5-10元杂芳基的取代基取代,其中a为0、1或2;
且,
R和R’各自独立地选自氢、C1-6烷基、3-8元环烷基、5-8元脂杂环基、6-10元芳基和5-10元杂芳基;任选地,其中所述C1-6烷基、3-8元环烷基、5-8元脂杂环基、6-10元芳基、5-10元杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、C1-4烷基、卤代C1-4烷基、羟基C1-4烷基、RS(O)2-、C1-4烷氧基、3-8元环烷基、5-8元脂杂环基、6-10元芳基和5-10元杂芳基的取代基取代,或者,R和R’与所连的N原子一起形成4-10元脂杂环基。
在本申请的某些优选实施方案中,所述式I化合物中R5选自氢、氧基、卤素、C1-10烷基、3-20元环烷基、3-20元脂杂环基、5-10元杂芳基、RO-、RC(O)-、ROC(O)-、RR’N-、RR’NC(O)-、RS(O)a-和RR’NSO2-;任选地,其中所述C1-10烷基、3-20元环烷基、3-20元脂杂环基、5-10元杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素(优选为氟)、羟基、氨基、氰基、C1-6烷基、C1-6烷氧基和3-8元环烷基的取代基取代,其中a为0、1或2;
其中,R和R’各自独立地选自氢、C1-6烷基和3-8元环烷基;任选地,其中所述C1-6烷基和3-8元环烷基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、C1-4烷基、C1-4烷氧基和C1-4烷基磺酰基的取代基取代。
在本申请的某些优选实施方案中,所述式I化合物中R5选自氢、氧基、卤素、C1-6烷基、3-8元环烷基、3-8元脂杂环基、5-6元杂芳基、RO-、RC(O)-、ROC(O)-、RR’N-、RR’NC(O)-、RS(O)a-和RR’NSO2-;任选地,其中所述C1-6烷基、3-8元环烷基、3-8元脂杂环基、5-6元杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素 (优选为氟)、羟基、氨基、氰基、C1-4烷基、C1-4烷氧基和3-6元环烷基的取代基取代,其中a为0、1或2;
其中,R和R’各自独立地选自氢、C1-4烷基和3-6元环烷基;任选地,其中所述C1-4烷基和3-6元环烷基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、C1-2烷基、C1-2烷氧基和C1-2烷基磺酰基的取代基取代。
在本申请的某些优选实施方案中,所述式I化合物中R5选自氢、氧基、卤素、C1-4烷基、3-6元环烷基、3-6元脂杂环基、5-6元杂芳基、RO-、RC(O)-、ROC(O)-、RR’N-、RR’NC(O)-、RS(O)a-和RR’NSO2-;任选地,其中所述C1-4烷基、3-6元环烷基、3-6元脂杂环基、5-6元杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素(优选为氟)、羟基、氨基、氰基、C1-2烷基、C1-2烷氧基和3-6元环烷基的取代基取代,其中a为0、1或2;
其中,R和R’各自独立地选自氢、C1-2烷基和3-6元环烷基;任选地,其中所述C1-2烷基和3-6元环烷基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、C1-2烷基、C1-2烷氧基和C1-2烷基磺酰基的取代基取代。
在本申请的某些优选实施方案中,所述式I化合物中R5选自氢、溴、羟甲基、1-羟基乙基、2-羟基-2-丙基、二氟甲基、三氟甲基、1,1-二氟乙基、五氟乙基、环丙基-二氟甲基、1-氨基乙基、环丙基甲基、环丙基、1-甲基4-吡唑基、1-甲基-3-吡唑基、2-甲基-[1.3.4]氧代二唑-5-基、3-甲基-[1.2.4]氧代二唑-5-基、甲氧基、环丙基氧基、乙酰基、乙氧羰基、环丙基氨基、氨酰基、甲氨酰基、N,N-二甲胺酰基、乙氨酰基、2-羟基乙基氨酰基、2-甲氧基乙基氨酰基、2-羟基异丁氨酰基、2-甲磺酰基乙基氨酰基、甲硫基和甲磺酰基。
在本申请的某些优选实施方案中,所述式I化合物中R6不存在或选自氢和卤代C1-10烷基(例如卤代C1-6烷基;例如卤代C1-4烷基;例如三氟甲基)。
在本申请的某些优选实施方案中,所述式I化合物中R6不存在或选自氢和三氟甲基。
在本申请的某些优选实施方案中,所述式I化合物中m=1,n=2;或者,m=2,n=2。
在本申请的某些优选实施方案中,所述化合物具有如式I-A所示的结构,
Figure PCTCN2016110007-appb-000008
其中,
A和B与相连的原子一起形成脂环、脂杂环、芳环或杂芳环,任选地,其中所述脂环、脂杂环、芳环或杂芳环各自独立地被一个或多个(例如1、2、3或4个)选自卤素、 羟基、氨基、氰基、羧基、硝基、烷基、卤代烷基、烯基、炔基、环烷基、脂杂环基、芳基、杂芳基、RO-、ROC(O)-、RC(O)O-、RC(O)-、RR’N-、RC(O)N(R”)-、RR’NC(O)-和RS(O)a的取代基取代,其中a为0、1或2;
环X为脂环、脂杂环、芳环或杂芳环,优选地,所述环X为含氮脂杂环或杂芳环;优选地,所述环X为五元或六元含氮脂杂环或杂芳环;优选地,所述环X为五元或六元含氮杂芳环;所述环X例如为吡咯环、咪唑环、吡唑环、三氮唑环或哌啶环;
m和n各自独立地选自0、1、2和3,且m+n=3;
R1、R2、R3和R4各自独立地选自氢、卤素、羟基、氨基、氰基、羧基、硝基、烷基、烯基、炔基、环烷基、脂杂环基、芳基、杂芳基、RO-、ROC(O)-、RC(O)O-、RC(O)-、RR’N-、RS(O)a-、RC(O)N(R’)-和RR’NC(O)-;任选地,其中所述烷基、烯基、炔基、环烷基、脂杂环基、芳基、杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、羧基、硝基、C1-4烷基、C1-4烷氧基、3-8元环烷基、5-8元脂杂环基、6-10元芳基、5-10元杂芳基和RSO2-的取代基取代;
R5和R6各自独立地选自不存在、氢、氧基、卤素、羟基、氨基、氰基、羧基、硝基、烷基、烯基、炔基、环烷基、脂杂环基、芳基、杂芳基、RO-、RC(O)-、ROC(O)-、RC(O)O-、RR’N-、RC(O)N(R”)-、RR’NC(O)-、ROC(O)N(R’)-、RR’NC(O)N(R”)-、RS(O)a-、RR’NSO2-和RS(O)2N(R’)-;任选地,其中所述烷基、烯基、炔基、环烷基、脂杂环基、芳基或杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、羧基、硝基、C1-4烷基、C1-4烷氧基、3-8元环烷基、5-8元脂杂环基、6-10元芳基、5-10元杂芳基和RSO2-的取代基取代,其中a为0、1或2;或者,
R5或R6并在与之相连的环X上形成多环系统,其中,所述R5或R6选自3-8元脂环、5-8元脂杂环、6-10元芳环和5-10元杂芳环;
R、R’和R”各自独立地选自氢、C1-10烷基、3-20元环烷基、5-20元脂杂环基、6-20元芳基和5-20元杂芳基;任选地,其中所述C1-10烷基、3-20元环烷基、5-20元脂杂环基、6-20元芳基、5-20元杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、羧基、硝基、C1-4烷基、卤代C1-4烷基、羟基C1-4烷基、C1-4烷氧基、3-8元环烷基、5-8元脂杂环基、6-10元芳基、5-10元杂芳基和RSO2-的取代基取代。
在本申请的某些优选实施方案中,所述式I-A化合物的限制条件为,所述环X不为吡咯烷环、2-吡咯烷酮环、吗啉环、3-吗啉酮环、四氢呋喃环、2-噁唑烷酮环、四氢噻吩环和噻吩烷砜环。
在本申请的某些优选实施方案中,所述式I-A化合物中A和B与相连的原子一起形成芳环(例如6-20元芳环、6-14元芳环或6-10元芳环),其中,所述芳环未被取代或各自独立地被一个或多个(例如1、2、3或4个)选自卤素(例如氟、氯、溴或碘)、羟基、氨基、氰基、羧基、硝基和C1-10烷基的取代基取代。
在本申请的某些优选实施方案中,所述式I-A化合物中A和B与相连的原子一起 形成苯环,其中,所述苯环未被取代或各自独立地被一个或多个(例如1、2、3或4个)选自卤素(例如氟)的取代基取代。
在本申请的某些优选实施方案中,所述式I-A化合物中R1、R2、R3和R4各自独立地选自氢、卤素、羟基、氨基、氰基、羧基、硝基、C1-10烷基和卤代C1-10烷基。
在本申请的某些优选实施方案中,所述式I-A化合物中R1、R2、R3和R4各自独立地选自氢、卤素、羟基、氨基、氰基、C1-6烷基(例如甲基)和卤代C1-6烷基(例如三氟甲基)。
在本申请的某些优选实施方案中,所述式I-A化合物中R1、R2和R3为氢原子。
在本申请的某些优选实施方案中,所述式I-A化合物中R4为卤素。
在本申请的某些优选实施方案中,所述式I-A化合物中R4为氟。
在本申请的某些优选实施方案中,所述式I-A化合物中A和B与相连的原子一起形成苯环,R1、R2和R3为氢,且R4为为氟。
在本申请的某些优选实施方案中,所述式I-A化合物中R5选自氢、氧基、卤素、C1-10烷基、3-20元环烷基、3-20元脂杂环基、5-10元杂芳基、RO-、RC(O)-、ROC(O)-、RR’N-、RR’NC(O)-、RS(O)a-、RR’NSO2-,任选地,其中所述C1-10烷基、3-20元环烷基、3-20元脂杂环基、5-10元杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素(优选为氟)、羟基、氨基、氰基、C1-6烷基、C1-6烷氧基和3-8元环烷基的取代基取代,其中a为0、1或2;
其中,R和R’各自独立地选自氢、C1-6烷基和3-8元环烷基,任选地,其中所述C1-6烷基和3-8元环烷基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、C1-4烷基、C1-4烷氧基和C1-4烷基磺酰基的取代基取代。
在本申请的某些优选实施方案中,所述式I-A化合物中R5选自氢、卤素、C1-6烷基、3-12元环烷基、RC(O)-和RR’NC(O)-,任选地,其中所述C1-6烷基和3-12元环烷基各自独立地被一个或多个(例如1、2、3或4个)选自卤素(优选为氟)、羟基、氨基、氰基、C1-4烷基、C1-4烷氧基和3-8元环烷基的取代基取代;
其中,R和R’各自独立地选自氢、C1-6烷基和3-8元环烷基。
在本申请的某些优选实施方案中,所述式I-A化合物中R5选自氢、卤素、C1-4烷基、3-6元环烷基、RC(O)-和RR’NC(O)-,任选地,其中所述C1-4烷基各自独立地被一个或多个(例如1、2、3或4个)选自卤素(优选为氟)和羟基的取代基取代;
其中,R和R’各自独立地选自氢、C1-4烷基和3-6元环烷基。
在本申请的某些优选实施方案中,所述式I-A化合物中R5选自氢、甲基、三氟甲基、环丙基甲基、2-羟基-2-丙基、环丙基、环丙甲酰基、氨酰基、甲氨酰基和二甲氨酰基。
在本申请的某些优选实施方案中,所述式I-A化合物中R6不存在或选自氢和卤代C1-10烷基。
在本申请的某些优选实施方案中,所述式I-A化合物中R6不存在或为氢。
在本申请的某些优选实施方案中,所述式I-A化合物中R6不存在。
在本申请的某些优选实施方案中,所述式I-A化合物中R6为氢。
在本申请的某些优选实施方案中,所述式I-A化合物中环X为五元杂芳环且杂原子为氮、或六元含氮脂杂环;例如吡唑环或哌啶环。
在本申请的某些优选实施方案中,所述式I-A中m=1,n=2;或者,m=2,n=2。
在本申请的某些优选实施方案中,所述式I-A化合物中
Figure PCTCN2016110007-appb-000009
选自如下结构:
Figure PCTCN2016110007-appb-000010
在本申请的某些优选实施方案中,所述式I-A化合物中多环系统选自如下结构:
Figure PCTCN2016110007-appb-000011
在本申请的某些实施方案中,所述多环系统可以具有两个手性中心(手性原子以*符号标示),本领域技术人员应当理解,本申请的化合物包含其所有构型,例如(R,R),(R,S),(S,S)或(S,R)构型。
在本申请的某些实施方案中,所述化合物具有式I-B所示结构,
Figure PCTCN2016110007-appb-000012
其中,
A和B与相连的原子一起形成脂环、脂杂环、芳环或杂芳环;任选地,其中所述脂环、脂杂环、芳环或杂芳环各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、羧基、硝基、烷基、卤代烷基、烯基、炔基、环烷基、脂杂环基、芳基、杂芳基、RO-、ROC(O)-、RC(O)O-、RC(O)-、RR’N-、RC(O)N(R”)-、RR’NC(O)-和RS(O)a-的取代基取代,其中a为0、1或2;
环X为脂环、脂杂环、芳环或杂芳环;优选地,所述环X为含氮脂杂环或杂芳环;优选地,所述环X为五元或六元含氮脂杂环或杂芳环;更优选地,所述环X为五元或六元含氮杂芳环;例如为吡咯环、咪唑环、吡唑环、三氮唑环或吡啶酮环;
m和n各自独立地选自0、1、2和3,且m+n=3或4;
R1、R2、R3和R4各自独立地选自氢、卤素、羟基、氨基、氰基、羧基、硝基、烷基、烯基、炔基、环烷基、脂杂环基、芳基、杂芳基、RO-、ROC(O)-、RC(O)O-、RC(O)-、RR’N-、RS(O)a-、RC(O)N(R”)-和RR’NC(O)-;任选地,其中所述烷基、烯基、炔基、环烷基、脂杂环基、芳基、杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、羧基、硝基、C1-4烷基、C1-4烷氧基、3-8元环烷基、5-8元脂杂环基、6-10元芳基、5-10元杂芳基和RSO2-的取代基取代,其中a为0、1或2;
R5和R6各自独立地选自不存在、氢、氧基、卤素、羟基、氨基、氰基、羧基、硝基、烷基、烯基、炔基、环烷基、脂杂环基、芳基、杂芳基、RO-、RC(O)-、ROC(O)-、RC(O)O-、RR’N-、RC(O)N(R”)-、RR’NC(O)-、ROC(O)N(R’)-、RR’NC(O)N(R”)-、RS(O)a-、RR’NSO2-和RS(O)2N(R’)-;任选地,其中所述烷基、烯基、炔基、环烷基、脂杂环基、芳基或杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、羧基、硝基、C1-4烷基、C1-4烷氧基、3-8元环烷基、5-8元脂杂环基、6-10元芳基、5-10元杂芳基和RSO2-的取代基取代,其中a为0、1或2;或者,
R5或R6并在与之相连的环X上形成多环系统,其中,所述R5或R6选自3-8元脂环、5-8元脂杂环、6-10元芳环和5-10元杂芳环;
R、R’和R”各自独立地选自氢、C1-10烷基、3-20元环烷基、5-20元脂杂环基、6-20元芳基和5-20元杂芳基;任选地,其中所述C1-10烷基、3-20元环烷基、5-20元脂杂环基、6-20元芳基或5-20元杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、羧基、硝基、C1-4烷基、卤代C1-4烷基、羟基C1-4烷基、C1-4烷氧基、3-8元环烷基、5-8元脂杂环基、6-10元芳基、5-10元杂芳基和RSO2-的取代基取代。
在本申请的某些优选实施方案中,所述式I-B化合物的限制性条件为,
当A和B与相连的碳原子形成未取代的苯环,R1、R2和R3均为H,R4为F时,
1)且当式I-B中
Figure PCTCN2016110007-appb-000013
Figure PCTCN2016110007-appb-000014
时,R6不存在,R5不为H、羟基、甲基、异丙基、环丙基、三氟甲基、二氟甲基和三氟乙基;
2)且当式I-B中
Figure PCTCN2016110007-appb-000015
Figure PCTCN2016110007-appb-000016
时,R5和R6不全为H、且当R5或R6其中一个为H时,另一个不为三氟甲基和乙氧羰基;
3)且式I-B中
Figure PCTCN2016110007-appb-000017
不为
Figure PCTCN2016110007-appb-000018
4)所述环X不为吡咯烷环、2-吡咯烷酮环、吗啉环、3-吗啉酮环和2-噁唑烷酮环;并且,
5)所述式I-B化合物不为:4-{[3-(3,4-二氢-1H-吡咯并[1,2-a]吡嗪-2-羰基)-4-氟-苯基]甲基}-2H-酞嗪-1-酮、4-{[3-(6,8-二氢-5H-[1,2,4]三唑并[1,5-a]吡嗪-7-羰基)-4-氟-苯基]甲基}-2H-酞嗪-1-酮、4-{[4-氟-3-(2-三氟甲基-6,8-二氢-5H-[1,2,4]三唑并[1,5-a]吡嗪-7-羰基)-苯基]甲基}-2H-酞嗪-1-酮、和4-[4-氟-3-(5,6-二氢-8H-咪唑并[1,5-a]吡嗪-7-羰基)苄基]-2H-酞嗪-1-酮。
在本申请的某些优选实施方案中,所述式I-B化合物中A和B与相连的原子一起形成芳环(例如6-20元芳环、6-14元芳环或6-10元芳环),其中,所述芳环未被取代或各自独立地被一个或多个(例如1、2、3或4个)选自卤素(例如氟、氯、溴或碘)、羟基、氨基、氰基、羧基、硝基和C1-10烷基的取代基取代。
在本申请的某些优选实施方案中,所述式I-B化合物中A和B与相连的原子一起形成苯环,其中,所述苯环未被取代或各自独立地被一个或多个(例如1、2、3或4个)选自卤素(例如氟)的取代基取代。
在本申请的某些优选实施方案中,所述式I-B化合物中R1、R2、R3和R4各自独立地选自氢、卤素、羟基、氨基、氰基、羧基、硝基、C1-10烷基和卤代C1-10烷基。
在本申请的某些优选实施方案中,所述式I-B化合物中R1、R2、R3和R4各自独立地选自氢、卤素、羟基、氨基、氰基、C1-6烷基(例如甲基)和卤代C1-6烷基(例如三氟甲基)。
在本申请的某些优选实施方案中,所述式I-B化合物中R1、R2和R3为氢原子。
在本申请的某些优选实施方案中,所述式I-B化合物中R4为卤素。
在本申请的某些优选实施方案中,所述式I-B化合物中R4为氟。
在本申请的某些优选实施方案中,所述式I-B化合物中A和B与相连的原子一起形成苯环,R1、R2和R3为氢,且R4为为氟。
在本申请的某些优选实施方案中,所述式I-B化合物中R5选自氢、氧基、卤素、C1-10烷基、3-20元环烷基、3-20元脂杂环基、5-10元杂芳基、RO-、RC(O)-、ROC(O)-、RR’N-、RR’NC(O)-、RS(O)a-、RR’NSO2-,任选地,其中所述C1-10烷基、3-20元环烷基、3-20元脂杂环基、5-10元杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素(优选为氟)、羟基、氨基、氰基、C1-6烷基、C1-6烷氧基和3-8元环烷基的取代基取代,其中a为0、1或2;
其中,R和R’各自独立地选自氢、C1-6烷基和3-8元环烷基,任选地,其中所述 C1-6烷基和3-8元环烷基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、C1-4烷基、C1-4烷氧基和C1-4烷基磺酰基的取代基取代。
在本申请的某些优选实施方案中,所述式I-B化合物中R5选自氢、氧基、卤素、C1-6烷基、3-12元环烷基、3-12元脂杂环基、5-6元杂芳基、RO-、RC(O)-、ROC(O)-、RR’N-、RR’NC(O)-和RS(O)a-,任选地,其中所述C1-6烷基、3-12元环烷基、3-12元脂杂环基、5-6元杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素(优选为氟)、羟基、氨基、氰基、C1-4烷基、C1-4烷氧基和3-8元环烷基的取代基取代,其中a为0、1或2;
其中,R和R’各自独立地选自氢、C1-4烷基和3-6元环烷基,任选地,其中所述C1-4烷基和3-6元环烷基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、C1-4烷基、C1-4烷氧基和C1-4烷基磺酰基的取代基取代。
在本申请的某些优选实施方案中,所述式I-B化合物中R5选自氢、氧基、卤素、C1-4烷基、3-6元环烷基、3-6元脂杂环基、5-6元杂芳基、RO-、RC(O)-、ROC(O)-、RR’N-、RR’NC(O)-和RS(O)a-,任选地,其中所述C1-4烷基、3-6元环烷基、3-6元脂杂环基、5-6元杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素(优选为氟)、羟基、氨基和3-6元环烷基的取代基取代,其中a为0、1或2;
其中,R和R’各自独立地选自氢、C1-4烷基和3-6元环烷基,任选地,其中所述C1-4烷基和3-6元环烷基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、C1-4烷基、C1-4烷氧基和C1-4烷基磺酰基的取代基取代。
在本申请的某些优选实施方案中,所述式I-B化合物中R5选自氢、溴、羟甲基、2-羟基-2-丙基、1-羟基乙基、二氟代甲基、三氟甲基、1,1-二氟乙基、五氟乙基、1-氨基乙基、环丙基甲基、环丙基二氟甲基、环丙基、1-甲基-1H-吡唑-4-基、1-甲基-1H-吡唑-3-基、3-甲基-[1,2,4]-噁二唑-5-基、5-甲基-[1,3,4]-噁二唑-2-基、甲氧基、环丙基氧基、乙氧羰基、环丙基氨基、氨酰基、甲氨酰基、乙氨酰基、N,N-二甲胺酰基、2-羟基乙氨酰基、2-甲氧基乙氨酰基、2-羟基异丁基氨酰基、2-甲磺酰基乙氨酰基、甲硫基和甲磺酰基。
在本申请的某些优选实施方案中,所述式I-B化合物中R6不存在或选自氢和卤代C1-10烷基。
在本申请的某些优选实施方案中,所述式I-B化合物中R6不存在或选自氢和卤代C1-6烷基。
在本申请的某些优选实施方案中,所述式I-B化合物中R6不存在。
在本申请的某些优选实施方案中,所述式I-B化合物中R6选自氢和卤代C1-4烷基(例如三氟甲基)。
在本申请的某些优选实施方案中,所述式I-B中m=1,n=2;或者,m=2,n=2。
在本申请的某些优选实施方案中,所述式I-B中环X为五元杂芳环且杂原子为氮、 或六元含氮脂杂环,优选地,所述
Figure PCTCN2016110007-appb-000019
选自以下结构:
Figure PCTCN2016110007-appb-000020
在本申请的某些优选实施方案中,本申请第一方面任一项所述的氢为氕(H)或氘(D)。
在本申请的某些优选实施方案中,所述多环系统选自如下结构:
Figure PCTCN2016110007-appb-000021
Figure PCTCN2016110007-appb-000022
本申请第一方面任一项所述的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型,其中所述化合物选自:
4-[4-氟-3-(2,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)苄基]-2H-酞嗪-1-酮(化合物1);
4-[4-氟-3-(2,4,6,7-四氢-吡唑并[4,3-c]吡啶-5-羰基)苄基]-2H-酞嗪-1-酮(化合物2);
4-[4-氟-3-(6,7-二氢-4H-[1,2,3]三氮唑并[1,5-a]吡嗪-5-羰基)苄基]-2H-酞嗪-1-酮(化合物3);
N-甲基-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-2-甲酰胺(化合物4);
N-甲基-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺(化合物5);
4-[3-(2-环丙甲酰-2,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物6);
4-[3-(2-环丙基甲基-2,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物7);
4-[4-氟-3-(2-甲基-2,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)-苄基]-2H-酞嗪-1-酮(化合物8);
4-[3-(2-环丙甲酰基-2,4,6,7-四氢-吡唑并[4,3-c]吡啶-5-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物9);
顺-4-[4-氟-3-(4,5,5a,6,7,8,9,9a-八氢-[1,2,4]三氮唑并[4,3-a][1,6]萘啶-7-羰基)-苄基]-2H-酞嗪-1-酮(化合物10);
反-4-[4-氟-3-(4,5,5a,6,7,8,9,9a-八氢-[1,2,4]三氮唑并[4,3-a][1,6]萘啶-7-羰基)-苄基]-2H-酞嗪-1-酮(化合物11);
4-[3-(1-环丙基-1,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物12);
7-[2-氟-5-(4-氧代-3,4-二氢-1-酞嗪基)甲基-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺(化合物13);
N,N-二甲基-7-[2-氟-5-(4-氧代-3,4-二氢-1-酞嗪基)甲基-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺(化合物14);
7-[2-氟-5-(4-氧代-3,4-二氢-1-酞嗪基甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-羧酸乙酯(化合物15);
4-[4-氟-3-(3-三氟甲基-1,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)苄基]-2H-酞嗪-1-酮(化合物16);
N-甲基-7-[2-氟-5-(4-氧代-3,4-二氢-1-酞嗪基)甲基苯甲酰基]-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-3-甲酰胺(化合物17);
7-[2-氟-5-(4-氧杂-3,4-二氢-1-酞嗪基)甲基苯甲酰基]-2-三氟甲基-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-3-羧酸乙酯(化合物18);
4-[3-(2-环丙基-2,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)-4-氟苯基]-2H-酞嗪-1-酮(化合物19);
N-甲基-6-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)苯甲酰基]-4,5,6,7-四氢-2H-吡唑[3,4-c]吡啶-3-甲酰胺(化合物20);
6-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-4,5,6,7-四氢-1H-吡唑[3,4-c]吡啶-3-甲酰胺(化合物21);
4-[4-氟-3-(2-羟甲基-6,7-二氢-4H-吡唑[1,5-a]吡嗪-5-羰基)-苄基]-2H-酞嗪-1-酮(化 合物22);
4-[3-(3-溴-6,7-二氢-4H-[1,2,3]三氮唑并[1,5-a]吡嗪-5-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物23);
N-(2-羟乙基)-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺(化合物24);
N,N-二甲基-6-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-4,5,6,7-四氢-1H-吡唑并[3,4-c]吡啶-3-甲酰胺(化合物25);
4-[4-氟-3-(6-氧代-1,3,4,6-四氢-吡啶并[1,2-a]吡嗪-2-羰基)苄基]-2H-酞嗪-1-酮(化合物26);
4-[3-(3-溴-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物27);
4-[3-(3-溴-6,7,8,9-四氢-5H-[1,2,4]三氮唑并[4,3-d][1,4]二氮杂卓-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物28);
4-{4-氟-3-[3-(1-羟基-1-甲基-乙基)-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮(化合物29);
N-甲基-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-2-三氟甲基-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-3-甲酰胺(化合物30);
N-乙基-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-2-三氟甲基-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-3-甲酰胺(化合物31);
4-{4-氟-3-[3-(1-羟基-1-甲基-乙基)-2-三氟甲基-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮(化合物32);
4-[4-氟-3-(3-甲氧基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-苄基]-2H-酞嗪-1-酮(化合物33);
N-(2-甲氧基乙基)-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺(化合物34);
N-(2-羟基-2-甲基丙基)-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺(化合物35);
N-(2-甲基磺酰基乙基)-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺(化合物36);
4-[3-(3-乙酰基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物37);
4-{4-氟-3-[3-(1-甲基-1H-吡唑-4-基)-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮(化合物38);
4-{4-氟-3-[3-(1-甲基-1H-吡唑-4-基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮(化合物39);
4-[4-氟-3-(3-甲巯基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-苄基]-2H-酞嗪-1-酮(化合物40);
4-[4-氟-3-(3-甲基磺酰基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-苄基]-2H-酞嗪-1-酮(化合物41);
4-{4-氟-3-[3-(2-羟基丙基-2-基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮(化合物42);
4-{4-氟-3-[3-(2-羟基丙基-2-基)-1,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基]-苄基}-2H-酞嗪-1-酮(化合物43);
4-{4-氟-3-[3-(1-羟基乙基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮(化合物44);
4-{4-氟-[(3-(1-甲基-1H-吡唑-3-基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮(化合物45);
4-(3-(3-(1,1-二氟乙基)-5,6-二氢-8H-[1,2,4]三氮唑[4,3-a]吡嗪-7-羰基)-4-氟苄基)酞嗪-1(2H)-酮(化合物46);
4-[3-(3-环丙基-1,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物47);
4-[3-(3-环丙基-5,6-二氢-8H-咪唑并[1,5-a]吡嗪-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物48);
4-[3-(3-环丙基-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物49);
4-[3-(3-环丙基-2-三氟甲基-5,6二氢-8H-咪唑并[1,2-a]吡嗪-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物50);
4-{4-氟-3-[3-(3-甲基-[1,2,4]-噁二唑-5-基)-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮(化合物51);
4-{4-氟-3-[3-(5-甲基-[1,3,4]-噁二唑-2-基)-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮(化合物52);
4-[3-(3-环丙基氨基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物53);
4-[3-(3-环丙基甲基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物54);
4-{3-[3-(环丙基二氟甲基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物55);
4-[3-(3-环丙基-6,7-二氢-4H-[1,2,3]三氮唑并[1,5-a]吡嗪-5-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物56);
4-[3-(3-环丙氧基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-4-氟苄基]-2H-酞嗪 -1-酮(化合物57);
4-{3-[3-(1-氨基乙基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基]-4-氟苄基}-2H-酞嗪-1-酮(化合物58);
4-[3-(3-环丙基-1-三氟甲基-5,6-二氢-8H-咪唑并[1,5-a]吡嗪-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物59);
4-{3-[3-(1,1-二氟乙基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基]-4-氟苄基}-7-氟-2H-酞嗪-1-酮(化合物60);
4-[3-(3-环丙基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-4-氟苄基]-7-氟-2H-酞嗪-1-酮(化合物61);
4-[3-(3-二氟甲基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-4-氟苄基]-7-氟-2H-酞嗪-1-酮(化合物62);
4-[4-氟-3-(3-全氟乙基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)苄基]-2H-酞嗪-1-酮(化合物63);
7-氟-4-[4-氟-3-(3-五氟乙基-5,6,7,8-四氢-[1,2,4]三唑[4,3-a]吡嗪-7-羰基)苄基]-2H-酞嗪-1-酮(化合物64);和
4-[4-氟-3-(6-甲基-3-五氟乙基-5,6,7,8-四氢-[1,2,4]三唑[4,3-a]吡嗪-7-羰基)苄基]-2H-酞嗪-1-酮(化合物65)。
本申请第二方面提供本申请第一方面任一项所述的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型的制备方法,其选自以下方法:
(1)当式I中R5为RR’NC(O)-时,所述化合物可通过以下路线合成:
Figure PCTCN2016110007-appb-000023
将化合物A与化合物B’进行缩合反应得到化合物C;将化合物C与化合物RR’NH进行氨酯交换得到化合物D;
(2)当式I中R5为RC(O)-时,所述化合物可通过以下路线合成:
Figure PCTCN2016110007-appb-000024
将化合物A与化合物B”进行缩合反应得到化合物C’;将化合物C’与化合物RCO-Lg进行酰基化反应得到化合物D’;或者,
(3)当式I中R5和R6为本申请第一方面所述的其它基团时,所述化合物可通过以下路线合成:
Figure PCTCN2016110007-appb-000025
将化合物A与化合物B进行缩合反应得到式I化合物;
其中,RCO-Lg中的Lg表示离去基团,例如卤素、-OTs、-OCOR等,其余各原子和取代基的定义如本申请第一方面所述。
在本申请的某些优选实施方案中,所述缩合反应的操作如下:将0.8-1.2当量的化合物A和1.0当量的化合物B、化合物B’或化合物B”(游离胺或盐,例如盐酸盐)溶于溶剂(例如DMF或THF),室温下加入0.8-1.2当量的缩合剂(例如HATU,EDCI,T3P等)和2.0-5.0当量的碱(例如DIPEA,Et3N,吡啶等),反应结束后向反应液加入水和有机溶剂(例如乙酸乙酯或甲基叔丁基醚),分液,萃取,干燥和旋干,粗品经制备薄层色谱纯化或柱层析纯化得到目标产物。
化合物B为市售可得或通过本领域常规实验方法制备得到。在本申请的某些优选实施方案中,化合物B的合成包括但不限于以下方法:
(1)将化合物B-1与Lg-R5-R5进行亲核取代反应得化合物B-2;化合物B-2脱氨基保护基得到化合物B:
Figure PCTCN2016110007-appb-000026
或者,
Figure PCTCN2016110007-appb-000027
(2)将化合物B-3与化合物B-4进行Suzuki反应得到化合物B-2;化合物B-2脱氨基保护基得到化合物B:
Figure PCTCN2016110007-appb-000028
或者,
(3)将化合物B-5进行加成/还原反应得到化合物B-2;化合物B-2脱氨基保护基得到化合物B:
Figure PCTCN2016110007-appb-000029
其中,Lg为亲核取代反应的离去基团,例如为卤素、-OTs、-OCOR等;P为常见的氨基保护基,例如Boc、Fmoc、Cbz、Alloc、Pht、Tos、Dmb、Bn等;R5’为可经一步加成/还原反应得到本申请所述的R5的取代基,例如酯基、酰基等,所述加成/还原反应的试剂例如为格氏试剂、NaBH4、NaBH3CN等,其余各原子或取代基定义如本申请第一方面所述。
在本申请的某些优选实施方案中,氨基的脱保护可采用本领域公知方法,对此本申请不做限制。
在本申请的某些优选实施方案中,所述化合物B可通过以下方法合成:
方法一:
Figure PCTCN2016110007-appb-000030
将中间体1和碱(例如三乙胺,DIPEA,碳酸钾等)溶于溶剂(例如二氯甲烷,四氢呋喃,DMF等),低温或室温下滴加酰氯的溶液(溶剂可采用二氯甲烷,四氢呋喃,乙氰等)反应结束后,萃取,分液,纯化,得到中间体2-1和2-2,将所得中间体脱除保 护基后得到目标中间体;
方法二:
Figure PCTCN2016110007-appb-000031
将中间体1溶于溶剂(例如乙醚,四氢呋喃等),然后在低温或室温下(-80℃-室温)加入碱(例如NaH,t-BuOK,LDA,LiHMDS等)并在此温度下反应10-30分钟后,滴加卤代烃的溶液(例如乙醚,四氢呋喃等)。反应结束后经过萃取,干燥纯化可以得到中间体3-1和3-2,将所得中间体脱除保护基后得到目标中间体;
方法三:
Figure PCTCN2016110007-appb-000032
将中间体1溶于溶剂(例如甲苯,二氧六环,DMF等)然后加入碱(例如碳酸钠,磷酸钾,碳酸钾等),所得混合物在惰性气体保护下加入钯催化剂(例如Pd2(dba)3,Pd(OAc)2,Pd(dppf)Cl2等)和配体(例如dppf,xantphos,x-phos等),反应可在室温或者加热(60-150℃)的条件下进行也可以在微波反应器中加热进行,反应结束后,经过分液,萃取以及纯化可以得到中间体4-1和4-2,将所得中间体脱除保护基后可以得到目标中间体。
方法四:
Figure PCTCN2016110007-appb-000033
将SM-1溶于溶剂(例如四氯化碳,氯仿,二氯甲烷等)然后在室温或低温下加入溴代试剂(例如溴素,NBS等)得到溴代化合物Int-1。该化合物与2-氨基吡嗪溶于溶剂(例如THF,甲醇,乙醇等)加入碱(例如碳酸钾,三乙胺等)在60-120℃下反应。反应结 束后经过后处理和纯化可以得到化合物Int-2。该中间体溶于溶剂(例如甲醇,乙醇,异丙醇等)催化氢化可以得到带酯基的目标产物,该反应也可以加入酸(例如醋酸,盐酸等)以加速或者促使反应进行;
化合物Int-2与胺溶于溶剂(例如THF,甲醇,乙醇,DMF,DMSO等)在室温或者升温(60-150℃)加碱(例如三乙胺,DIPEA等)或者不加碱的条件下进行胺解,纯化后可以得到化合物Int-3,该反应也可以采用无溶剂的方式进行也可以在微波反应器中进行,Int-3在溶剂(例如甲醇,乙醇,异丙醇等)中进行催化氢化可以得到酰胺类目标中间体,该反应也可以加入酸(例如醋酸,盐酸等)以加速或者促使反应进行;
方法五:
Figure PCTCN2016110007-appb-000034
起始原料吡嗪-2-肼和碱(例如三乙胺,DIPEA,等)溶于溶剂(例如二氯甲烷,THF,乙氰等)然后在低温或者室温下加入草酰氯单酯,反应完成后经过后处理和纯化得到Int-4,将该化合物溶于溶剂(甲醇,乙醇,DMF等)在加热的条件下(40-150℃)反应,可加碱(例如甲醇钠,乙醇钠,叔丁醇钠等)或者不加碱。反应完成后经后处理和纯化得到关环的产物,该产物在溶剂(例如甲醇,乙醇,异丙醇等)中进行催化氢化可以得到一类带酯基的目标产物,该反应也可以加入酸(例如醋酸,盐酸等)以加速或者促使反应进行;
Int-5与各种胺溶于溶剂(例如THF,甲醇,乙醇,DMF,DMSO等)在室温或者升温(60-150℃)条件下进行胺解,可加碱(例如三乙胺,DIPEA等)或者不加碱,纯化后可以得到Int-6。该反应也可以采用无溶剂的方式进行也可以在微波反应器中进行,Int-6在溶剂(例如甲醇,乙醇,异丙醇等)中进行催化氢化可以得到一类酰胺类的目标中间体,该反应也可以加入酸(例如醋酸,盐酸等)以加速或者促使反应进行。
方法六:
Figure PCTCN2016110007-appb-000035
将N保护的3-哌啶酮溶于溶剂(例如THF,乙醚等)然后在低温(-90-0℃)或者室温下加入碱(例如KHMDS、LiHMDS、LDA等)并在此温度下反应30分钟后,加入酯,反应结束后经过后处理和纯化得到1,3-二酮类衍生物,将此衍生物溶于溶剂(例如 DCM,THF,乙醇等)与肼的水溶液,醇溶液或者盐酸盐反应可关环得到中间体吡唑衍生物,该中间体脱除保护基后可得目标中间体。
方法七:
当式I中R5并在与之相连的环X上形成多环系统时,化合物B可通过以下方法合成:
Figure PCTCN2016110007-appb-000036
将起始原料溶于溶剂(例如甲苯,THF等)然后加入硫代试剂(例如劳森试剂,P2S5等)常温或升温条件下反应,结束后经过纯化可以得到羰基被硫代的中间体,将该中间体与酰基肼溶于适当的溶剂(例如甲醇,乙醇等),然后加入碱(例如三乙胺,DIPEA,甲醇钠等)在室温或者升温(40-120℃)或者在微波下反应,该反应也可加入等当量的醋酸汞,氯化汞类汞盐或者碳酸银类的银盐以加速或者促进反应的进行,反应完全后,经后处理和纯化后可以得到关环三氮唑中间体,脱除保护基后可得目标中间体。
方法八:
Figure PCTCN2016110007-appb-000037
起始原料吡嗪-2-肼和取代的二氟乙酸(SM-2)溶于强酸(例如多聚磷酸等)然后在加热条件下(例如90-150℃)合环,反应完成后经过后处理和纯化得到中间体INT-1,该产物溶于溶剂(例如甲醇,乙醇,异丙醇等)使用催化剂(例如钯碳、氢氧化钯炭等)在氢气的环境下进行催化氢化可以得到目标中间体INT-2,该反应也可以加入酸(例如醋酸,盐酸等)以加速或者促使反应进行;
或者,可以用起始原料吡嗪-2-肼和取代的二氟乙酸酐(SM-3)在溶剂中(例如二氯甲烷、四氢呋喃等)缩合得到酰肼中间体INT-3。该产物溶于强酸(例如多聚磷酸/polyphosphoric acid等)然后在加热条件下(例如90-150℃)合环,反应完成后经过后处理和纯化得到中间体INT-1。
在另一个方面,本申请提供一种药物组合物,其包含本申请所述化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型,任选地,其还包含药学可接受的载体或赋形剂。
在本申请的某些优选实施方案中,所述化合物、其前体药物、代谢物形式、药学 上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型还可与一种或多种药物联用。因此,在本申请的某些优选实施方案中,所述药物组合物还包含一种或多种药物。在本申请的某些优选实施方案中,所述药物为抗肿瘤药物。在本申请的某些优选实施方案中,所述抗肿瘤药物选自替莫唑胺、阿霉素、紫杉醇、顺铂、卡铂、达卡巴嗪、拓扑替康、伊立替康、吉西他滨、贝伐单抗、anti-CTLA-4单抗Ipilimumab、anti-PD-1单抗pembrolizumab和Nivolumab以及anti-PD-L1单抗atezolizumab。
在本申请的某些优选实施方案中,所述载体包括但不限于:氧化铝,硬脂酸铝,卵磷脂,血清蛋白如人血清蛋白,磷酸盐,甘油,山梨酸,山梨酸钾,饱和植物脂肪酸的部分甘油酯混合物,水,硫酸鱼精蛋白,磷酸氢二钠,磷酸氢钾,氯化钠,锌盐,胶态氧化硅,三硅酸镁,聚乙烯吡咯烷酮,纤维素物质,聚乙二醇,羧甲基纤维素钠,聚丙烯酸酯,蜂蜡,羊毛脂。所述赋形剂是指在药物制剂中除主药以外的附加物。其性质稳定,与主药无配伍禁忌,不产生副作用,不影响疗效,在常温下不易变形、干裂、霉变、虫蛀、对人体无害、无生理作用,不与主药产生化学或物理作用,不影响主药的含量测定等。如片剂中的黏合剂、填充剂、崩解剂、润滑剂;中药丸剂中的酒、醋、药汁等;半固体制剂软膏剂、霜剂中的基质部分;液体制剂中的防腐剂、抗氧剂、矫味剂、芳香剂、助溶剂、乳化剂、增溶剂、渗透压调节剂、着色剂等均可称为赋形剂。
本申请的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或其药物组合物可以通过以下途径给药:胃肠外、局部、静脉内、口服、皮下、动脉内、真皮内、经皮、直肠、颅内、腹膜内、鼻内、肌内途径或作为吸入剂。
本申请的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或其药物组合物可根据给药途径配成各种适宜的剂型。
当口服用药时,本申请化合物可制成任意口服可接受的制剂形式,包括但不限于片剂、胶囊、水溶液或水悬浮液。其中,片剂使用的载体一般包括乳糖和玉米淀粉,另外也可加入润滑剂如硬脂酸镁。胶囊制剂使用的稀释剂一般包括乳糖和干燥玉米淀粉。水悬浮液制剂则通常是将活性成分与适宜的乳化剂和悬浮剂混合使用。任选地,以上口服制剂形式中还可加入一些甜味剂、芳香剂或着色剂。
当皮肤局部施用时,本申请化合物可制成适当的软膏、洗剂或霜剂等制剂形式,其中将活性成分悬浮或溶解于一种或多种载体中。软膏制剂可使用的载体包括但不限于:矿物油、液体凡士林、白凡士林、丙二醇、聚氧化乙烯、聚氧化丙烯、乳化蜡和水;洗剂或霜剂可使用的载体包括但不限于:矿物油、脱水山梨糖醇单硬脂酸酯、吐温60、十六烷酯蜡、十六碳烯芳醇、2-辛基十二烷醇、苄醇和水。
本申请化合物还可以无菌注射制剂形式用药,包括无菌注射水或油悬浮液或无菌 注射溶液,也可以是冻干形式。其中,可使用的载体和溶剂包括水、林格氏溶液和等渗氯化钠溶液。另外,灭菌的非挥发油也可用作溶剂或悬浮介质,如单甘油酯或二甘油酯。
本申请的药物制剂包括药学上可实施的任何制剂,例如口服制剂、肠胃外给药制剂等。
本申请的药物组合物和药物制剂可以含有0.01-2000mg的本申请化合物,优选含有0.1-1000mg本申请化合物,优选含有1-800mg本申请化合物,更优选含有10-600mg本申请化合物,特别优选含有50-500mg本申请化合物。
在本申请的某些优选实施方案中,进行合适的体外或体内测定来确定本申请组合物的效果以及给药是否适用于治疗个体所患疾病或医学疾病状态。这些测定的实例在下文非限制性实施例结合具体疾病或医学治疗进行了描述。通常,足以实现预防或治疗效果的本申请组合物的有效量为约0.001mg/千克体重/天至约10,000mg/千克体重/天。合适的情况下,剂量为约0.01mg/千克体重/天至约1000mg/千克体重/天。剂量范围可以为每天、每两天或每三天约0.01至1000mg/kg宿主体重,更通常为0.1至500mg/kg宿主体重。示例性的治疗方案为每两天一次或每周一次或每月一次给药。通常多次给予所述试剂,单次剂量之间的间隔可以是每天、每周、每月或每年。或者,可以以缓释制剂的形式给予所述试剂,在这种情况下,需要较少的给药频率。剂量和频率根据试剂在受试者中的半衰期而不同。也可以根据是预防性处理还是治疗性处理而不同。在预防性应用中,以相对低频率的间隔长期给予相对低的剂量。在治疗性应用中,有时需要以相对短的间隔给予相对高的剂量,直至疾病的进展被延缓或停止,并优选地直至个体表现出疾病症状的部分或完全改善,在此之后,可以给予患者预防方案。
在另一个方面,本申请提供所述化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药物组合物在制备抑制PARP的试剂中的用途。
在本申请的某些优选实施方案中,所述试剂为抑制PARP-1的试剂。
在另一个方面,本申请还提供一种抑制PARP活性的方法,所述方法包括向有需要的细胞施用有效量的本申请所述的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药物组合物。
在本申请的某些优选实施方案中,所述方法用于抑制PARP-1活性。
在本申请的某些优选实施方案中,所述方法用于抑制细胞中PARP-1活性。
在本申请的某些优选实施方案中,所述细胞为细胞系或来自受试者的细胞。
在另一个方面,本申请提供所述化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药物组合物在制备辅助治疗肿瘤的试剂或是用于增强放射或化学治疗效果的药物中的用途。
在另一个方面,本申请提供所述化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药物组合物,其用于辅助治疗肿瘤或是用于增强放射或化学治疗效果。
在另一个方面,本申请提供一种辅助治疗肿瘤或增强放射或化学治疗效果的方法,所述方法包括向有需要的受试者施用有效量的本申请所述的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药物组合物。
在另一个方面,本申请提供所述化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药物组合物在制备治疗肿瘤的药物中的用途。
在另一个方面,本申请提供所述化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药物组合物,其用于治疗肿瘤。
在另一个方面,本申请提供一种治疗肿瘤的方法,所述方法包括向有需要的受试者提供有效量的本申请所述的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药物组合物。
在另一个方面,本申请提供所述化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药物组合物在制备治疗血管疾病、神经退变性疾病或神经系统炎症的药物中的用途。
在另一个方面,本申请提供所述化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药物组合物,其用于治疗血管疾病、神经退变性疾病或神经系统炎症。
在另一个方面,本申请提供一种治疗血管疾病、神经退变性疾病或神经系统炎症的方法,所述方法包括向有需要的受试者施用有效量的本申请所述的化合物、其前体 药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药物组合物。
在另一个方面,本申请提供所述化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药物组合物,其用于抑制细胞中PARP活性。
在本申请的某些优选实施方案中,所述化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药物组合物用于抑制细胞中PARP-1活性。
在本申请的某些优选实施方案中,所述细胞为细胞系或来自受试者的细胞。
在本申请的某些优选实施方案中,所述化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药物组合物用于体内方法中。
在本申请的某些优选实施方案中,所述化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药物组合物用于体外方法中。
在另一个方面,本申请还提供一种抑制细胞中PARP活性的方法,所述方法包括给予细胞有效量的本申请所述的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药物组合物。
在本申请的某些优选实施方案中,所述方法用于抑制PARP-1活性。
在本申请的某些优选实施方案中,所述细胞为细胞系或来自受试者的细胞。
在本申请的某些优选实施方案中,所述方法在体内进行。
在本申请的某些优选实施方案中,所述方法在体外进行。
在另一个方面,本申请还提供所述化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药物组合物在制备试剂中的用途,所述试剂作为化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型辅助抑制肿瘤细胞增殖的试剂或是用于增强放射或化学方法抑制肿瘤细胞增殖的效果。
在本申请的某些优选实施方案中,所述肿瘤细胞为肿瘤细胞系或来自受试者的肿瘤细胞。
在本申请的某些优选实施方案中,所述试剂用于体内方法中。
在本申请的某些优选实施方案中,所述试剂用于体外方法中。
在另一个方面,本申请还提供所述化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药物组合物,其用于辅助抑制肿瘤细胞增殖或用于增强放射或化学方法抑制肿瘤细胞增殖的效果。
在本申请的某些优选实施方案中,所述肿瘤细胞为肿瘤细胞系或来自受试者的肿瘤细胞。
在本申请的某些优选实施方案中,所述化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请药物组合物用于体内方法中。
在本申请的某些优选实施方案中,所述化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药物用于体外方法中。
在另一个方面,本申请还提供一种辅助抑制肿瘤细胞增殖或增强放射或化学方法抑制肿瘤细胞增殖的效果的方法,所述方法包括给予肿瘤细胞有效量的本申请所述的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药物组合物。
在本申请的某些优选实施方案中,所述肿瘤细胞为肿瘤细胞系或来自受试者的肿瘤细胞。
在本申请的某些优选实施方案中,所述方法在体外进行。
在本申请的某些优选实施方案中,所述方法在体内进行。
在另一个方面,本申请还提供所述化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药物组合物在制备试剂中的用途,所述试剂用于抑制肿瘤细胞增殖。
在本申请的某些优选实施方案中,所述试剂用于体内方法中。
在本申请的某些优选实施方案中,所述试剂用于体外方法中。
在另一个方面,本申请还提供所述化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药物组合物,其用于抑制肿瘤细胞增殖。
在本申请的某些优选实施方案中,所述肿瘤细胞为肿瘤细胞系或来自受试者的肿瘤细胞。
在本申请的某些优选实施方案中,所述化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药 物组合物用于体内方法中。
在本申请的某些优选实施方案中,所述化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药物组合物用于体外方法中。
在另一个方面,本申请还提供一种抑制肿瘤细胞增殖的方法,所述方法包括给予细胞有效量的本申请所述的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型、或本申请所述的药物组合物。
在本申请的某些优选实施方案中,所述肿瘤选自乳腺癌、卵巢癌、结直肠癌、黑色素瘤、肺癌、胃肠道间质瘤、脑癌、宫颈癌、胰腺癌、前列腺癌、胃癌、慢性髓样白细胞过多症、肝癌、淋巴瘤、腹膜癌、软组织肉瘤、神经内分泌肿瘤和胶质母细胞瘤。
在本申请的某些优选实施方案中,所述肿瘤细胞选自乳腺癌细胞、卵巢癌细胞、结直肠癌细胞、黑色素瘤细胞、肺癌细胞、胃肠道间质瘤细胞、脑癌细胞、宫颈癌细胞、胰腺癌细胞、前列腺癌细胞、胃癌细胞、慢性髓样白细胞、肝癌细胞、淋巴瘤细胞、腹膜癌细胞、软组织肉瘤细胞、神经内分泌肿瘤细胞和胶质母细胞瘤细胞。
在本申请的某些优选实施方案中,所述肿瘤细胞为肿瘤细胞系或来自受试者的肿瘤细胞。
本申请所述的肿瘤包括恶性和良性肿瘤,相应地,所述肿瘤细胞包括恶性和良性肿瘤细胞。
本申请所述受试者为哺乳动物,例如牛科动物、马科动物、羊科动物、猪科动物、犬科动物、猫科动物、啮齿类动物、灵长类动物;其中,特别优选的受试者为人。
以下对本申请的术语进行解释,对于特定的术语,如果本申请中的含义与本领域技术人员通常理解的含义不一致,则以本申请中的含义为准;如果在本申请中没有定义,则其具有本领域技术人员通常理解的含义。除非有相反陈述,本申请中使用的术语具有下述含义。
本文所用术语“氢”及所述各基团中的氢,其包括氕(H),氘(D),氚(T)。
本文所用术语“烷基”是指直链或支链饱和烃基,例如C1-10烷基、C1-6烷基或C1-4烷基,非限制性实施例包括甲基、乙基、丙基、异丙基、丁基、异丁基、仲丁基、叔丁基、戊基、己基等。所述烷基未被取代,或被取代基进一步取代形成取代烷基,所述取代基可选自卤素、羟基、烷氧基、环烷基、脂杂环基、芳基或杂芳基。取代烷基的非限制性实施例包括卤代C1-4烷基、羟甲基、2-羟乙基、2-羟基丙-2-基、3-羟丙基、甲氧甲基、2-甲氧乙基、3-甲氧丙基、乙氧甲基、2-乙氧乙基、3-乙氧丙基、环丙基甲基、环丁基甲基、 环戊基甲基、环己基甲基、2-环丙基乙基、2-环丁基乙基、2-环戊基乙基、2-环己基乙基、3-环丙基丙基、3-环丁基丙基、3-环戊基丙基、3-环己基丙基、苄基、吡啶-2-甲基等。
本文所用术语“烯基”是指含有至少一个碳碳双键的直链或支链烃基,例如C2-10烯基、C2-6烯基或C2-4烯基,非限制性实施例包括乙烯基、丙烯基、丁烯基、2-甲基丙烯基、戊烯基、2-甲基丁烯基、3-甲基丁烯基、己烯基、2-甲基戊烯基、3-甲基戊烯基、4-甲基戊烯基、2-乙基丁烯基等。
本文所用术语“炔基”是指含有至少一个碳碳叁键的直链或支链烃基,例如C2-10炔基、C2-6炔基或C2-4炔基,非限制性实施例包括乙炔基、丙炔基、丁炔基、戊炔基、3-甲基丁炔基、己炔基、3-甲基戊炔基等。
本文所用术语“环烷基”是指饱和或部分不饱和的单环或多环环状烃基,例如包含3-20个碳原子的环烷基,例如包含3-12个碳原子的环烷基,例如包含3-10个碳原子的环烷基,例如包含3-8个碳原子的环烷基,例如包含3-6个碳原子的环烷基,例如包含5-8个碳原子的环烷基。单环烷基的非限制性实施例包括环丙基、环丁基、环戊基、环己基、环己烯基、环己二烯基、环庚基、环庚三烯基、环辛基等。多环环烷基包括螺环、稠环和桥环的环烷基。
本文所用术语“脂杂环基”是指包含至少一个选自N、O和S的杂原子的饱和或部分饱和的环状烃基。例如包含3-20、3-12、3-8、3-6、5-20、5-12、5-8或5-6个环成员的脂杂环基。脂杂环基的非限制性实施例包括吡咯烷基、哌啶基、哌嗪基、吗啉基、硫吗啉基、高哌嗪基等。
本文所用术语“芳基”是指具有芳香性的单环或稠合多环(例如双环或三环)基团,优选6-20元芳基,优选6-14元芳基,更优选6-10元芳基,芳基的非限制性实施例包括但不限于苯基和萘基等。
本文所用术语“杂芳基”是指被至少一个选自N、O或S的杂原子取代的5-20元芳香杂环基团,优选为5-10元,优选为5-6元杂芳基。杂芳基的非限制性实施例包括呋喃基、噻吩基、吡咯基、咪唑基、噁唑基、噻唑基、三氮唑基、四氮唑基、吡啶基、哒嗪基、嘧啶基和吡嗪基等。
本文所用术语“脂环”是指饱和或部分饱和的不具有芳香特性的碳环。例如包括3-20元脂环、3-12元脂环、3-8元脂环、4元脂环、5元脂环、6元脂环、7元脂环等。
本文所用术语“脂杂环”是指至少一个环成员为选自N、O和S的杂原子的脂肪族环状基团。例如含有1个或2个N原子的脂杂环、例如只含一个O原子的脂杂环、例如只含一个S原子的脂杂环等。例如包括3-20元脂杂环、3-12元脂杂环、3-8元脂杂环、4元脂杂环、5元脂杂环、6元脂杂环、7元脂杂环等。
本文所用术语“芳环”是指所有环成员均为碳原子的芳香环。例如包括6-20元芳环、6-14元芳环、6-10元芳环等。
本文所用术语“杂芳环”是指至少一个环成员为选自N、O和S的杂原子的具有芳 香性的环状基团。例如含有1个或2个N原子的杂芳环、例如只含一个O原子的杂芳环、例如只含一个S原子的杂芳环等。例如包括5-20元杂芳环、5-14元杂芳环、5-10元杂芳环、5-6元杂芳环等。
本文所用术语“卤素”包括氟、氯、溴和碘。
本文中“RO-”的非限制性实施例包括甲氧基、乙氧基、丙氧基和环丙氧基等。
本文中“ROC(O)-”的非限制性实施例包括甲氧羰基、乙氧羰基、丙氧羰基、异丙氧羰基和叔丁氧羰基等。
本文中“RC(O)O-”的非限制性实施例包括甲酰氧基、乙酰氧基和丙酰氧基等。
本文中“RC(O)-”的非限制性实施例包括甲酰基、乙酰基、丙酰基和环丙甲酰基等。
本文中“RR’N-”的非限制性实施例包括甲氨基、二甲胺基、乙氨基和二乙胺基等。
本文中“RC(O)N(R”)-”的非限制性实施例包括甲酰氨基、乙酰氨基和丙酰胺基等。
本文中“RR’NC(O)-”的非限制性实施例包括甲氨酰基、N,N-二甲氨酰基、乙氨酰基、2-羟乙基氨酰基、2-甲氧乙基氨酰基、2-羟基异丁氨酰基、2-甲磺酰基乙基氨酰基等。
本文中“RS(O)a-”包括取代的磺酰基、取代的亚磺酰基以及取代的巯基,其中取代的磺酰基的非限制性实施例包括甲磺酰基、乙磺酰基、苯磺酰基、对甲苯磺酰基等。取代的亚磺酰基的非限制性实施例包括甲基亚磺酰基、乙基亚磺酰基、苯基亚磺酰基、对甲苯基亚磺酰基等。取代的巯基的非限制性实施例包括甲巯基、苯巯基等。
本文中“RR’NS(O)2-”是指取代的氨基磺酰基,RR’NS(O)2-的非限制性实施例包括二甲氨基磺酰基等。
本文中“RS(O)2N(R’)-”是指取代的磺酰胺基,例如烷基磺酰胺基和芳基磺酰胺基等。RS(O)2N(R’)-的非限制性实施例包括甲磺酰胺基、乙磺酰氨基、苯磺酰胺基和对甲苯磺酰胺基等。
本文中的“氧基”特指
Figure PCTCN2016110007-appb-000038
所述氧基可取代在环烷基、脂杂环基、芳基、杂芳基等基团上的碳原子上形成羰基或以烯醇式存在,例如形成吡咯烷酮、吡啶-2-酮、酞嗪-1-酮等结构。
本领域技术人员应当理解,当本申请所述化合物的分子结构中存在共轭体系时,符合价键理论的共振结构也在本申请的保护范围之内,例如,
Figure PCTCN2016110007-appb-000039
式a、式b和式c均在本申请的保护范围内。
本文所用术语“异构体”包括本申请通式I化合物的所有可能的异构体形式,例如对映异构体、非对映异构体、差向异构体、顺反异构体、构象异构体等。例如,R和S构型的对映异构体、以及Z和E构型的顺反异构体等均在本申请的保护范围内。
本文所用术语“室温”也称为常温或一般温度,一般指室内温度,优选20℃-38℃, 例如20℃-30℃,例如20℃-25℃。
本申请式Ⅰ化合物或其药学可接受的盐还可以形成溶剂合物,例如水合物、醇合物等。本申请式Ⅰ化合物或其药学可接受的盐还可以以晶体存在,所述晶体是指构成化合物的分子、原子或离子在空间按一定规律周期重复的排列,其排列具有三维空间的周期性,隔一定的距离重复出现。化合物均可以两种或多种结晶状态存在,结构相同的分子结晶成不同的固体形式,称为“polymorph”,即为多晶型物或多晶形等。当涉及具体结晶形式或多晶型时,统称“crystal form”,即为本申请中使用的术语“晶型”,本申请包含所述式I化合物或其药学可接受的盐的任意晶型。
本文所用术语“有效量”是指足以实现所需预防和/或治疗效果的量,例如,实现预防或减轻与待治疗疾病相关的症状的量。
本文所用的术语“治疗”是指治疗性处理和预防性措施,其目的是预防或延缓(减轻)所针对的疾病状态或病症。如果受试者按照本文所述方法接受了治疗量的化合物、或其异构体、溶剂合物、药学可接受的盐或其药物组合物,该受试者一种或多种指征和症状表现出可观察到的和/或可检测出的降低或改善,则受试者被成功地“治疗”了。还应当理解,所述的疾病状态或病症的预防或治疗的不仅包括完全地预防或治疗,还包括未达到完全地预防或治疗,但实现了一些生物学或医学相关的结果。
本文所用术语“血管疾病”主要是指心肌缺血/再灌注损伤,损伤后各种形式的心脏衰竭,心肌病,循环性休克,心血管衰老,糖尿病的心血管并发症,心肌肥大,动脉粥样硬化,血管重塑,血管生成。
本文所用术语“神经退变性疾和神经系统炎症”主要是指由于氧化,亚硝化应激的有害影响而导致的中风,脑外伤,神经变性疾病(帕金森氏病,阿尔茨海默氏病和肌萎缩性侧索硬化)和神经系统发炎如多发性硬化等。
发明的有益效果
本申请通过对酞嗪酮类PARP抑制剂的构效关系进行深入研究,提供了一类酞嗪酮类化合物,所述化合物能实现下述的至少一种技术效果:
(1)能够明显提高对肿瘤细胞的增殖抑制作用;
(2)能够增加分子体内稳定性,降低产生毒性代谢物的可能性;
(3)能降低药物分子的毒性,使其在疾病治疗方面的安全性得到了进一步的提高,扩展了此类药物的适用病群范围;
(4)通过对酞嗪酮类化合物的结构修饰,减低了化合物在体内P450细胞色素酶系作用下的氧化代谢能力,提高了生物利用率;
(5)本申请的化合物具有良好的生物利用度、抑制有效性和安全性;
(6)本申请的化合物具有优良的长效性,由此可以减少给药次数,提高患者的依从性。
附图说明
图1为本申请实施例19产物的NOE图谱。
具体实施方式
下面将结合实施例对本申请的实施方案进行详细描述,但是本领域技术人员将会理解,下列实施例仅用于说明本申请,而不应视为限定本申请的范围。
化合物的结构是通过核磁共振(1HNMR)或质谱(MS)来确定的。
实施例中使用的核磁共振(NMR)图谱中的缩写示于以下:
s:单峰(singlet)、d:二重峰(doublet)、t:三重峰(triplet)、q:四重峰(quartet)、AB:双二重峰(double doublet)、m:多重峰(multiplet)、br:宽峰(broad);
1HNMR的测定是用JEOL Eclipse 400核磁仪,测定溶剂为氘代甲醇(CD3OD)、氘代氯仿(CDCl3),六氘代二甲基亚砜(DMSO-d6),内标为四甲基硅烷(TMS),化学位移是以10-6(ppm)作为单位给出。
MS的测定用Agilent(ESI)质谱仪,生产商:Agilent,型号:Agilent 6120B。
制备高效液相使用岛津LC-8A制备液相色谱仪(YMC,ODS,250×20mml色谱柱)。
薄层色谱硅胶板(TLC)使用Merck产的铝板(20×20cm),薄层层析分离纯化采用的规格是烟台产GF254(0.4-0.5nm)。
反应的监测采用薄层色谱法(TLC)或LCMS,使用的展开剂体系有:二氯甲烷和甲醇体系,正己烷和乙酸乙酯体系,石油醚和乙酸乙酯体系,溶剂的体积比根据化合物的极性不同而进行调节或者加入三乙胺等进行调节。
微波反应使用Biotage Initiator+(400W,RT-300℃)微波反应器。
柱层析一般使用青岛海洋200-300目硅胶为载体。洗脱剂的体系包括:二氯甲烷和甲醇体系,正己烷和乙酸乙酯体系,溶剂的体积比根据化合物的极性不同而进行调节,也可以加入少量的三乙胺进行调节。
实施例中无特殊说明,反应的温度为室温(20℃-30℃)实施例中未注明具体条件者,按照常规条件或制造商建议的条件进行。所用试剂或仪器未注明生产厂商者,均为可以通过市购获得的常规产品。
本申请所使用的试剂购自Acros Organics,Aldrich Chemical Company,特伯化学等公司。
在常规的合成法以及实施例、和中间体合成例中,各缩写的意思如以下所示:
DCM:二氯甲烷;
DIPEA:N,N-二异丙基乙胺;
DMAP:4-二甲氨基吡啶;
NOE:Nuclear Overhauser Effect,奥弗豪塞尔核效应;
MeOH:甲醇;
THF:四氢呋喃;
EtOH:乙醇;
DMF:N,N-二甲基甲酰胺;
NMP:N-甲基吡咯烷酮;
HATU:2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯;
LCMS:液相质谱联用仪;
NaH:钠氢;
TLC:薄层色谱;
T3P:丙基磷酸酐。
实施例1:
4-[4-氟-3-(2,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)苄基]-2H-酞嗪-1-酮(化合物1)
Figure PCTCN2016110007-appb-000040
反应路线:
Figure PCTCN2016110007-appb-000041
将化合物5-(3,4-二氢-4-氧代-1-酞嗪基)甲基-2-氟苯甲酸(50mg,0.17mmol),HATU(70mg,0.18mmol)和二异丙基乙基胺(0.58mL,3.5mmol)溶于DMF(5mL),室温下搅拌30分钟后加入4,5,6,7-四氢-2H-吡唑并[3,4-c]吡啶(22mg,0.18mmol),室温搅拌至LCMS显示原料消耗完全。将反应液倒入饱和氯化铵水溶液中,乙酸乙酯萃取。有机相用无水硫酸钠干燥,浓缩后残余物制备薄层硅胶板纯化得到目标产物(59mg,收率:86.1%)。
MS m/z(ESI):404[M+H]+
1HNMR(400MHz,CD3OD)δ:8.38-8.34(m,1H),7.97-7.93(m,1H),7.93-7.81(m,2H),7.52-7.36(m,3H),7.20-7.13(m,1H),4.90(m,1H),4.39-4.37(m,3H),3.49-3.48(t,1H,J=2Hz),3.30(m,1H),2.78-2.75(t,1H,J=6Hz),2.53(br s,1H).
实施例2:
4-[4-氟-3-(2,4,6,7-四氢-吡唑并[4,3-c]吡啶-5-羰基)苄基]-2H-酞嗪-1-酮(化合物2)
Figure PCTCN2016110007-appb-000042
标题化合物采用实施例1的方法合成,用4,5,6,7-四氢-2H-吡咯并[4,3-c]吡啶取代实施例1中所用的4,5,6,7-四氢-2H-吡唑并[3,4-c]吡啶。
MS m/z(ESI):404[M+H]+.
1HNMR(400MHz,CD3OD)δ:8.39-8.34(m,1H),8.00-7.78(m,3H),7.55-7.31(m,2H),7.28-7.10(m,2H),5.00-4.82(s,1H),4.78(s,1H),4.44-4.30(m,2H),3.85-3.51(m,1H),3.40-3.23(m,1H),2.82-2.79(m,1H),2.72-2.53(m,1H).
实施例3:
4-[4-氟-3-(6,7-二氢-4H-[1,2,3]三氮唑并[1,5-a]吡嗪-5-羰基)苄基]-2H-酞嗪-1-酮(化合物3)
Figure PCTCN2016110007-appb-000043
标题化合物采用实施例1的方法合成,用4,5,6,7-四氢-[1,2,3]三氮唑并[1,5-a]吡嗪取代实施例1中所用的4,5,6,7-四氢-2H-吡唑并[3,4-c]吡啶。
MS m/z(ESI):405[M+H]+.
1HNMR(400MHz,CDCl3)δ:8.47-8.41(m,1H),7.82-7.70(m,4H),7.67-7.65(m,1H),7.57(s,1H),7.32-7.25(m,1H),4.72(s,2H),4.34(s,2H),3.93(t,2H,J=4Hz),3.72(t,2H,J=4Hz).
实施例4:
N-甲基-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-2-甲酰胺(化合物4)
Figure PCTCN2016110007-appb-000044
第一步:化合物7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-2-甲酸甲酯
采用实施例1的方法合成,用5,6,7,8-四氢-咪唑并[1,2-a]吡嗪-2-羧酸甲酯取代实施例1中所用的4,5,6,7-四氢-2H-吡唑并[3,4-c]吡啶。
第二步:化合物N-甲基-7-[2-氟-5-(4-氧代-3,4-二氢酞嗪-1-甲基)苯甲酰基)-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-2-甲酰胺
将第一步的化合物7-[2-氟-5-(4-氧代-3,4二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-2-甲酸甲酯(100mg,0.21mmol)溶于甲胺的甲醇溶液(30%,5mL),将所得溶液在85℃下反应过夜。TLC监测反应完全后,将反应液浓缩,残余物倒入水中,二氯甲烷萃取。有机相用无水硫酸钠干燥,浓缩后制备薄层色谱纯化得到20mg目标产物,收率:20%。
MS m/z(ESI):461[M+H]+。
1HNMR(400MHz,CD3OD)δ:8.38-8.34(m,1H),8.00-7.78(m,3H),7.59-7.42(m,3H),7.23-7.15(m,1H),4.91(s,1H),4.58(s,1H),4.44-4.35(d,2H,J=4Hz),4.10-3.98(m,1H),3.80-3.68(m,1H),3.40-3.25(m,2H),2.96-2.78(m,3H).
实施例5:
N-甲基-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺(化合物5)
Figure PCTCN2016110007-appb-000045
反应路线:
Figure PCTCN2016110007-appb-000046
第一步:化合物7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酸乙酯
采用实施例1的方法合成,用5,6,7,8-四氢-[1,2,4]三氮唑并[4,3-a]哌嗪-3-甲酸乙酯取代实施例1中所用的4,5,6,7-四氢-2H-吡唑并[3,4-c]吡啶。
第二步:化合物N-甲基-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺
采用实施例4第二步的方法合成,用第一步的7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酸乙酯取代实施例4中所用的 7-[2-氟-5(4-氧代-3,4-二氢-酞嗪1-甲基)-苯甲酰基]-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-2-甲酸甲酯。
MS m/z(ESI):462[M+H]+.
1HNMR(400MHz,DMSO-d6)δ:12.60(br s,1H),8.97-8.82(m,1H),8.34-8.19(m,1H),8.04-7.78(m,3H),7.57-7.42(m,2H),7.35-7.22(m,1H),5.13-4.84(br s,1H),4.66(s,1H),4.41-4.23(m,4H),3.70-3.58(m,1H),3.56-3.40(m,1H),2.78-2.76(d,3H,J=8Hz).
实施例6:
4-[3-(2-环丙甲酰-2,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物6)
Figure PCTCN2016110007-appb-000047
反应路线:
Figure PCTCN2016110007-appb-000048
第一步:4-[4-氟-3-(2,4,5,6-四氢-吡唑并[3,4-c]吡啶-6-羰基)-苄基]-2H-酞嗪基-1-酮
采用实施例1的方法合成,用4,5,6,7-四氢-2H-吡唑并[3,4-c]吡啶取代实施例1中所用的4,5,6,7-四氢-2H-吡唑并[3,4-c]吡啶。
第二步:4-[3-(2-环丙甲酰基-2,4,5,6-四氢-吡唑并[3,4-c]吡啶-6-羰基)-4-氟苄基]-2H-酞嗪-1-酮
将第一步的4-[4-氟-3-(2,4,5,6-四氢-吡唑并[3,4-c]吡啶-6-羰基)-苄基]-2H-酞嗪基-1-酮(30mg,0.074mmol)和三乙胺(22.42mg,0.222mmol)溶于四氢呋喃(2mL),冰浴下滴加环丙基甲酰氯(11.6mg,0.111mmol)。10分钟后反应结束,冰水淬灭反应。水相用乙酸乙酯萃取,有机层用无水硫酸钠干燥,浓缩。残余物用制备薄层色谱纯化得到目标产物,收率:57%。
MS m/z(ESI):472[M+H]+.
1HNMR(400MHz,DMSO-d6)δ:12.66(br s,1H),8.29-8.19(m,2H),8.02-7.77(m,3H),7.50-7.19(m,3H),5.03-4.29(m,4H),3.48-3.36(m,2H),3.14-2.95(m,1H),2.77-2.53(m,2H),1.20-1.03(m,4H).
实施例7:
4-[3-(2-环丙基甲基-2,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)-4-氟苄基]-2H-酞嗪-1-酮 (化合物7)
Figure PCTCN2016110007-appb-000049
反应路线:
Figure PCTCN2016110007-appb-000050
第一步:2-环丙基甲基-2,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-碳酸叔丁酯
NaH(256.8mg,6.42mmol)加入到2,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-碳酸叔丁酯(250mg,1.07mmol)的N,N-二甲基甲酰胺(5mL)溶液中,所得混合物在室温下反应1小时后加入环丙甲基溴(288.9mg,2.14mmol)。室温下反应6小时后,用水淬灭反应,二氯甲烷萃取。二氯甲烷层经无水硫酸钠干燥,浓缩得到标题化合物(150mg粗品),直接用于下一步反应。
第二步:2-环丙基甲基-2,4,5,7-四氢-吡唑并[3,4-c]吡啶盐酸盐
将第一步的2-环丙基甲基-2,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-碳酸叔丁酯放入反应瓶中,然后加入氯化氢的乙酸乙酯溶液(4N,3mL)。混合物在室温下搅拌过夜,将浓缩得到的产物直接用于下一步反应。
第三步:4-[3-(2-环丙基甲基-2,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)-4-氟苄基]-2H-酞嗪-1-酮
将5-[(3,4-二氢-4-氧代-1-酞嗪基)甲基]-2-氟苯甲酸(129.3mg,0.43mmol),第二步的2-环丙基甲基-2,4,5,7-四氢-吡唑并[3,4-c]吡啶盐酸盐(76.7mg,0.43mmol),HATU(179.8mg,0.47mmol)和DIPEA(332.8mg,2.58mmol)溶于DMF,室温搅拌过夜。反应完全后,向反应液中加入水和乙酸乙酯,分液,乙酸乙酯层经无水硫酸钠干燥后浓缩。残余物经制备薄层色谱纯化后得到目标产物(5mg,收率:2.5%)。
MS m/z(ESI):458[M+H]+.
1HNMR(400MHz,DMSO-d6)δ:12.65(br s,1H),8.30-8.18(m,2H),8.02-7.75(m,3H),7.50-7.20(m,3H),5.05-4.31(m,4H),3.48-3.36(m,4H),3.15(m,2H),1.43-1.38(m,1H),1.20-1.03(m,4H).
实施例8:
4-[4-氟-3-(2-甲基-2,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)-苄基]-2H-酞嗪-1-酮(化合物8)
Figure PCTCN2016110007-appb-000051
反应路线
Figure PCTCN2016110007-appb-000052
第一步:2-甲基-2,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-碳酸叔丁酯
NaH(108mg,2.70mmol)加入到2,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-碳酸叔丁酯(100mg,0.45mmol)的N,N-二甲基甲酰胺(5mL)溶液中,所得混合物在室温下反应1小时后加入碘甲烷(127.8mg,0.90mmol)。室温下反应7小时后,用水淬灭反应,二氯甲烷萃取。二氯甲烷层经无水硫酸钠干燥,浓缩残余物经制备薄层色谱纯化后得到产物(75mg,收率:70%)。
第二步:2-甲基-2,4,5,7-四氢-吡唑并[3,4-c]吡啶盐酸盐
将第一步的2-甲基-2,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-碳酸叔丁酯放入反应瓶中,然后加入氯化氢的乙酸乙酯溶液(4N,3mL)。混合物在室温下搅拌过夜,将浓缩得到的产物直接用于下一步反应。
第三步:4-[4-氟-3-(2-甲基-2,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)-苄基]-2H-酞嗪-1-酮
将5-[(3,4-二氢-4-氧代-1-酞嗪基)甲基]-2-氟苯甲酸(95.4mg,0.32mmol),第二步的2-甲基-2,4,5,7-四氢-吡唑并[3,4-c]吡啶盐酸盐(43mg,0.32mmol),HATU(146mg,0.38mmol)和DIPEA(206.4mg,1.60mmol)溶于DMF,室温搅拌过夜。反应完全后,向反应液中加入水和乙酸乙酯,分液,乙酸乙酯层经无水硫酸钠干燥后浓缩。残余物经制备薄层色谱纯化后得到目标产物(30mg,收率:23%)。
核磁图谱证明该样品是两个异构体的混合物,比例为3:1。
Figure PCTCN2016110007-appb-000053
MS m/z(ESI):418[M+H]+.
1HNMR(400MHz,DMSO-d6)δ:12.71(br s,1H),8.27-8.25(d,1H,J=8Hz),8.02-7.95(m,1H),7.93-7.80(m,2H),7.51-7.32(m,3H),7.31-7.18(m,1H),4.82-4.60(m,1H),4.41-4.22(m,3H),3.90-3.64(m,3H),3.39-3.29(m,2H),2.70-2.53(m,1H),2.46-2.38(m,1H).
实施例9:
4-[3-(2-环丙甲酰基-2,4,6,7-四氢-吡唑并[4,3-c]吡啶-5-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物9)
Figure PCTCN2016110007-appb-000054
采用实施例6第二步的方法合成,由4-[4-氟-3-(2,4,6,7-四氢-吡唑并[4,3-c]吡啶-5-羰基)-苄基]-2H-酞嗪-1-酮和环丙甲酰氯制备而成。
MS m/z(ESI):472[M+H]+.
1HNMR(400MHz,DMSO-d6)δ:12.69(br s,1H),8.30-8.25(m,1H),8.00-7.98(m,1H),7.96-7.83(m,2H),7.49-7.30(m,3H),7.29-7.18(m,1H),4.85-4.58(m,1H),4.40-4.25(m,3H),3.90-3.64(m,2H),3.39-3.29(m,2H),2.58-2.47(m,1H),1.15-1.00(m,4H).
实施例10:
顺-4-[4-氟-3-(4,5,5a,6,7,8,9,9a-八氢-[1,2,4]三氮唑并[4,3-a][1,6]萘啶-7-羰基)-苄基]-2H-酞嗪-1-酮(化合物10)
Figure PCTCN2016110007-appb-000055
反应路线:
Figure PCTCN2016110007-appb-000056
采用实施例1的方法合成,用顺-4,5,5a,6,7,8,9,9a-八氢-[1,2,4]三氮唑并[4,3-a][1,6]萘啶(合成参见ACS Med.Chem.Lett.(DOI:10.1021/ml500503n))取代实施例1中所用的4,5,6,7-四氢-2H-吡唑并[3,4-c]吡啶。
MS m/z(ESI):459[M+H]+.
1HNMR(400MHz,DMSO-d6)δ:12.68-12.55(m,1H),8.53-8.44(m,1H),8.32-8.20(m,1H),8.05-7.73(m,3H),7.63-7.16(m,3H),4.58-4.43(m,1H),4.40-4.27(m,3H),3.28-2.92(m,5H),2.87-2.72(m,1H),2.13-1.64(m,4H).
实施例11:
反-4-[4-氟-3-(4,5,5a,6,7,8,9,9a-八氢-[1,2,4]三氮唑并[4,3-a][1,6]萘啶-7-羰基)-苄基]-2H-酞嗪-1-酮(化合物11)
Figure PCTCN2016110007-appb-000057
反应路线:
Figure PCTCN2016110007-appb-000058
采用实施例1的实验步骤合成,用反-4,5,5a,6,7,8,9,9a-八氢-[1,2,4]三氮唑并[4,3-a][1,6]萘啶)合成参见ACS Med.Chem.Lett.(DOI:10.1021/ml500503n))取代实施例1中所用的4,5,6,7-四氢-2H-吡唑并[3,4-c]吡啶。
MS m/z(ESI):459[M+H]+。
1HNMR(400MHz,DMSO-d6)δ:12.61(br s,1H),8.53(d,1H,J=24Hz),8.31-8.23(m,1H),8.01-7.80(m,3H),7.53-7.30(m,2H),7.28-7.20(m,1H),4.75-4.59(m,1H),4.38(s,2H),3.96-3.81(m,1H),3.57-3.14(m,3H),3.11-2.58(m,4H),1.82-1.34(m,3H)。
实施例12:
4-[3-(1-环丙基-1,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物12)
Figure PCTCN2016110007-appb-000059
反应路线:
Figure PCTCN2016110007-appb-000060
第一步:1-环丙基-1,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-碳酸叔丁酯
将化合物1,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-碳酸叔丁酯(200mg,2.32mmol)完全溶解于5mL1,4-二氧六环,加入环丙基硼酸(250mg,1.12mmol),醋酸铜(200mg,1.10mmol),吡啶(87mg,1.10mmol)和DMAP(403.15mg,3.30mmol).将所得反应混合物在100℃下搅拌过夜至LCMS监测原料消耗完全后,以二氯甲烷萃取,饱和食盐水返洗,硫酸钠干燥,减压浓缩得到棕色油状物,残余物经制备薄层色谱纯化后得到目标产物(160mg,收率:26.2%)。环丙基的区域异构化学通过实施例19的1H核磁共振的NOE实验确定(环丙基的氢与吡唑氢无相关峰)。
第二步:1-环丙基-1,4,5,7-四氢-吡唑并[3,4-c]吡啶盐酸盐
将1-环丙基-1,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-碳酸叔丁酯(160mg,0.61mmol)溶于氯化氢的乙酸乙酯溶液(3mL),室温下搅拌过夜。反应液浓缩后得到的残余物直接用于下一步反应。
第三步:4-[3-(1-环丙基-1,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)-4-氟苄基]-2H-酞嗪-1-酮
采用实施例1的方法合成,用第二步的1-环丙基-1,4,5,7-四氢-吡唑并[3,4-c]吡啶盐酸盐取代实施例1中所用的4,5,6,7-四氢-2H-吡唑并[3,4-c]吡啶。
MS m/z(ESI):444[M+H]+
1HNMR(400MHz,CD3OD)δ:8.39-8.32(m,1H),8.02-7.95(m,1H),7.92-7.81(m,2H),7.64-7.55(m,2H),7.35-7.22(m,1H),5.21-5.02(m,1H),4.84-4.76(m,2H),4.43(s,2H),4.17-3.79(m,6H),2.28-2.09(m,2H),2.01-1.90(m,2H)。
实施例13:
7-[2-氟-5-(4-氧代-3,4-二氢-1-酞嗪基)甲基-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺(化合物13)
Figure PCTCN2016110007-appb-000061
标题化合物采用实施例4的方法合成,用实施例15的7-[2-氟-5-(4-氧代-3,4-二氢-1-酞嗪基)甲基-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酸乙酯取代实施例4中所用的7-[2-氟-5-(4-氧代-3,4-二氢-1-酞嗪基)甲基苯甲酰基]-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-2-甲酸甲酯,用7mol/L氨甲醇溶液取代实施例9中所用30%甲胺甲醇溶液。
MS m/z(ESI):448[M+H]+。
1HNMR(400MHz,DMSO-d6)δ:12.60(br s,1H),8.31-8.23(m,2H),8.01-7.80(m,4H),7.55-7.45(m,2H),7.32-7.24(m,1H),5.00(br s,1H),4.40-4.24(m,4H),4.13-4.05(m,1H),3.67-3.60(m,1H),3.17(d,2H,J=4Hz)。
实施例14:
N,N-二甲基-7-[2-氟-5-(4-氧代-3,4-二氢-1-酞嗪基)甲基-苯甲酰基]--5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺(化合物14)
Figure PCTCN2016110007-appb-000062
标题化合物采用实施例4的方法合成,用实施例15的7-[2-氟-5-(4-氧代-3,4-二氢-1-酞嗪基)甲基-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酸乙酯取代实施例4中所用的7-[2-氟-5-(4-氧代-3,4-二氢-1-酞嗪基)甲基-苯甲酰基]-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-2-甲酸甲酯,用二甲胺溶液取代实施例4中所用30%甲胺甲醇溶液。
MS m/z(ESI):476[M+H]+
1HNMR(400MHz,CDCl3)δ:8.45-8.41(m,1H),7.87-7.81(m,1H),7.75-7.69(m,3H),7.65-7.61(m,1H),7.28-7.22(m,1H),4.46(s,2H),4.34(s,2H),4.27(t,2H,J=4Hz),3.72(t,2H,J=4Hz),3.09(s,6H)。
实施例15:
7-[2-氟-5-(4-氧代-3,4-二氢-1-酞嗪基甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-羧酸乙酯(化合物15)
Figure PCTCN2016110007-appb-000063
标题化合物采用实施例1的方法合成,用5,6,7,8-四氢-[1,2,4]三氮唑并[4,3-a]吡嗪-3-羧酸乙酯取代实施例1中所用的4,5,6,7-四氢-2H-吡唑并[3,4-c]吡啶。
MS m/z(ESI):477[M+H]+。
1HNMR(400MHz,CD3OD)δ:8.39-8.32(m,1H),7.98-7.79(m,3H),7.56-7.43(m,2H),7.25-7.17(m,1H),5.12(s,1H),4.79(s,1H),4.51-4.30(m,6H),3.80-3.73(m,1H),1.45-1.34(m,3H).
实施例16:
4-[4-氟-3-(3-三氟甲基-1,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)苄基]-2H-酞嗪-1-酮(化合物16)
Figure PCTCN2016110007-appb-000064
标题化合物采用实施例1的方法合成,用3-三氟甲基-4,5,6,7-四氢-1H-吡唑并[3,4-c]哌啶取代实施例1中所用的4,5,6,7-四氢-2H-吡唑并[3,4-c]吡啶。
MS m/z(ESI):476[M+H]+
1HNMR(400MHz,CDCl3)δ:8.49-8.46(m,1H),7.78-7.71(m,3H),7.65-7.61(m,1H),7.59-7.56(m,1H),7.31-7.25(m,1H),4.72(s,2H),4.34(s,2H),3.94(t,2H,J=4Hz),3.16(t,2H,J=4Hz)。
实施例17:
N-甲基-7-[2-氟-5-(4-氧代-3,4-二氢-1-酞嗪基)甲基苯甲酰基]-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-3-甲酰胺(化合物17)
Figure PCTCN2016110007-appb-000065
标题化合物采用实施例4的方法合成,用中间体7-[2-氟-5-(4-氧代-3,4-二氢-1-酞嗪基)甲基苯甲酰基]-5,6-二氢-8H-咪唑[1,2-a]吡嗪-3-羧酸乙酯取代实施例4中所用的7-[2-氟-5(4-氧代-3,4-二氢-1-酞嗪基)甲基苯甲酰基]-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-2-甲酸甲酯。
MS m/z(ESI):461[M+H]+
1HNMR(400MHz,DMSO-d6)δ:12.59(br s,1H),8.29-8.20(m,2H),8.03-7.80(m,3H),7.59-7.43(m,3H),7.33-7.23(m,1H),4.83(brs,1H),4.48(br s,1H),4.37-4.29(m,3H),4.28-4.22(m,1H),3.66-3.59(m,1H),2.72(d,2H,J=4Hz)。
实施例18:
7-[2-氟-5-(4-氧杂-3,4-二氢-1-酞嗪基)甲基苯甲酰基]-2-三氟甲基-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-3-羧酸乙酯(化合物18)
Figure PCTCN2016110007-appb-000066
Figure PCTCN2016110007-appb-000067
标题化合物采用实施例1的实验步骤合成,用2-三氟甲基-5,6,7,8-四氢-咪唑并[1,2-a]吡嗪-3-羧酸乙酯取代实施例1中所用的4,5,6,7-四氢-2H-吡唑并[3,4-c]吡啶。
MS m/z(ESI):490[M+H]+
1HNMR(400MHz,CD3OD)δ:8.40-8.31(m,1H),8.00-7.78(m,3H),7.57-7.43(m,2H),7.26-7.13(m,1H),4.97(s,1H),4.64(s,1H),4.52-4.28(m,6H),3.80-3.67(m,2H),3.27-3.19(m,1H),1.38-1.33(m,3H)。
实施例19:
4-[3-(2-环丙基-2,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)-4-氟苯基]-2H-酞嗪-1-酮(化合物19)
Figure PCTCN2016110007-appb-000068
标题化合物采用实施例12的方法合成。环丙基的区域异构化学通过1H核磁共振的NOE实验确定(结果见图1)。
MS m/z(ESI):444[M+H]+
1HNMR(400MHz,DMSO-d6)δ:12.58(brs,1H),8.27-8.25(m,1H),7.99-7.96(m,1H),7.91-7.81(m,2H),7.55(s,1H),7.44-7.43(m,1H),7.37-7.35(m,1H),7.27-7.21(m,1H),4.64-4.61(m,1H),4.33-4.26(m,3H),3.84-3.57(m,2H),2.60-2.55(m,1H),2.45-2.40(m,2H),1.23(s,1H),1.00-0.85(m,4H)。
实施例20:
N-甲基-6-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)苯甲酰基]-4,5,6,7-四氢-2H-吡唑[3,4-c]吡啶-3-甲酰胺(化合物20)
Figure PCTCN2016110007-appb-000069
第一步:6-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)苯甲酰基]-4,5,6,7-四氢-2H-吡咯并[3,4-c]吡啶-3-羧酸乙酯
采用类似于实施例1的实验步骤合成,用4H-吡咯-3-羧酸乙酯取代实施例1中所用的4,5,6,7-四氢-2H-吡唑并[3,4-c]吡啶。
第二步:N-甲基-6-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)苯甲酰基]-4,5,6,7-四氢-2H-吡唑[3,4-c]吡啶-3-甲酰胺
采用类似于实施例4的实验步骤合成,用第一步的6-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)苯甲酰基]-4,5,6,7-四氢-2H-吡咯并[3,4-c]吡啶-3-羧酸乙酯取代实施例4中所用的7-[2-氟-5(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-2-甲酸甲酯。
MS m/z(ESI):461[M+H]+
1H NMR(400MHz,DMSO-d6)δ:13.02(br s,1H),12.60(br s,1H),8.27-8.25(m,1H),8.02-7.82(m,3H),7.46-7.41(m,2H),7.29-7.24(m,1H),4.82-4.70(m,1H),4.40-4.30(m,2H),3.45-3.38(m,3H),2.78-2.76(m,2H),2.71(d,2H,J=4Hz).
实施例21:
6-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-4,5,6,7-四氢-1H-吡唑[3,4-c]吡啶-3-甲酰胺(化合物21)
Figure PCTCN2016110007-appb-000070
第一步:6-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-4,5,6,7-四氢-1H-吡咯并[3,4-c]吡啶-3-羧酸乙酯
合成参见实施例20第一步。
第二步:6-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-4,5,6,7-四氢-1H-吡唑[3,4-c]吡啶-3-甲酰胺
采用类似于实施例4的实验步骤合成,用第一步的6-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-4,5,6,7-四氢-2H-吡咯并[3,4-c]吡啶-3-羧酸乙酯取代实施例4中所用的7-[2-氟-5(4-氧代-3,4-二氢-1-酞嗪-甲基)-苯甲酰基]-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-2-甲酸甲酯,用氨水取代实施例4中所用的甲胺溶液。
MS m/z(ESI):447[M+H]+
1HNMR(400MHz,CD3OD)δ:8.38-8.35(m,1H),7.99-7.81(m,3H),7.50-7.47(m,1H),7.40-7.38(m,1H),7.20-7.16(m,1H),5.35-5.33(m,1H),4.40-4.36(m,2H),3.57-3.28(m,3H),2.79-2.76(m,2H).
实施例22:
4-[4-氟-3-(2-羟甲基-6,7-二氢-4H-吡唑[1,5-a]吡嗪-5-羰基)-苄基]-2H-酞嗪-1-酮(化合物22)
Figure PCTCN2016110007-appb-000071
标题化合物采用实施例1的方法合成,用4,5,6,7-四氢-吡咯并[1,5-a]吡嗪-2-基)-甲醇取代实施例1中所用的4,5,6,7-四氢-2H-吡唑并[3,4-c]吡啶。
MS m/z(ESI):434[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.60(br s,1H),8.27(d,1H,J=8Hz),7.98(t,1H,J=8Hz),7.92-7.82(m,2H),7.50-7.38(m,2H),7.30-7.24(m,1H),6.12(s,0.6H),5.94(s,0.4H),5.02-4.97(m,1H),4.84(br s,1H),4.47(br s,1H),4.37-4.33(m,4H),4.10(br s,1H),3.99-3.96(m,1H),3.67-3.65(m,1H).
实施例23:
4-[3-(3-溴-6,7-二氢-4H-[1,2,3]三氮唑并[1,5-a]吡嗪-5-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物23)
Figure PCTCN2016110007-appb-000072
标题化合物采用实施例1的方法合成,用3-溴-4,5,6,7-四氢-[1,2,3]三氮唑并[1,5-a]吡嗪取代实施例1中所用的4,5,6,7-四氢-2H-吡唑并[3,4-c]吡啶。
MS m/z(ESI):483[M+H]+
1HNMR(400MHz,DMSO-d6)δ:8.38-8.34(m,1H),7.98-7.94(m,1H),7.90-7.80(m,2H),7.56-7.52(m,1H),7.48-7.44(m,1H),7.21(t,1H,J=9Hz),4.89(m,1H),4.54-4.50(m,1H),4.41-4.36(m,3H),3.83-3.79(m,1H),2.99(s,1H),2.86(s,1H).
实施例24:
N-(2-羟乙基)-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺(化合物24)
Figure PCTCN2016110007-appb-000073
第一步:7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酸乙酯
合成参见实施例5第一步方法。
第二步:N-(2-羟乙基)-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺
采用实施例4的方法合成,用第一步的7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酸乙酯取代实施例4中所用的7-[2-氟-5-(4-氧代-3,4二氢-1-酞嗪基)-甲基)-苯甲酰基]-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-2-甲酸甲酯,用乙醇胺取代实施例4中所用的甲胺溶液。
MS m/z(ESI):492[M+H]+
1HNMR(400MHz,DMSO-d6)δ:12.59(br s,1H),8.79-8.73(m,1H),8.26(d,1H,J=8Hz),7.97(t,1H,J=8Hz),7.93(t,1H,J=7Hz),7.84(t,1H,J=7Hz),7.53-7.46(m,2H),5.00(br s,1H),4.77(t,1H,J=6Hz),4.68-4.60(m,1H),4.40-4.25(m,4H),3.68-3.62(m,1H),3.54-3.48(m,2H),4.41-4.36(m,3H),3.83-3.79(m,1H),3.35-3.28(m,2H).
实施例25:
N,N-二甲基-6-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-4,5,6,7-四氢-1H-吡唑并[3,4-c]吡啶-3-甲酰胺(化合物25)
Figure PCTCN2016110007-appb-000074
第一步:6-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-4,5,6,7-四氢-2H-吡咯并[3,4-c]吡啶-3-羧酸乙酯
合成参见实施例20第一步。
第二步:N,N-二甲基-6-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-4,5,6,7-四氢-1H-吡唑并[3,4-c]吡啶-3-甲酰胺
采用实施例4方法合成,用第一步的6-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-4,5,6,7-四氢-2H-吡咯并[3,4-c]吡啶-3-羧酸乙酯取代实施例4中所用的7-[2-氟-5-(4-氧代-3,4-二氢-1-酞嗪基-甲基)-苯甲酰基]-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-2-甲酸甲酯,用二甲胺取代实施例4中所用的甲胺溶液。
MS m/z(ESI):475[M+H]+
1HNMR(400MHz,DMSO-d6)δ:13.02(br s,1H),12.59(br s,1H),8.28-8.25(m,1H),8.02-7.82(m,3H),7.46-7.41(m,2H),7.29-7.24(m,1H),4.82-4.70(m,1H), 4.40-4.32(m,2H),3.55-3.46(m,6H),2.78-2.76(m,2H),2.71(d,2H,J=4Hz).
实施例26:
4-[4-氟-3-(6-氧代-1,3,4,6-四氢-吡啶并[1,2-a]吡嗪-2-羰基)苄基]-2H-酞嗪-1-酮(化合物26)
Figure PCTCN2016110007-appb-000075
标题化合物采用实施例1的方法合成,用1,2,3,4-四氢-吡啶并[1,2-a]吡嗪-6-酮取代实施例1中所用的4,5,6,7-四氢-2H-吡唑并[3,4-c]吡啶。
MS m/z(ESI):431[M+H]+
1H NMR(400MHz,DMSO-d6)δ:8.39-8.35(m,1H),7.98-7.94(m,2H),7.90-7.80(m,2H),7.58-7.37(m,3H),7.24-7.16(m,1H),6.50(d,1H,J=9Hz),6.46-6.02(m,1H),4.85(br s,1.2H),4.44(br s,0.8H),4.42-4.37(m,3H),4.21(t,1H,J=5Hz),3.91(t,1H,J=6Hz),3.62(t,1.2H,J=5Hz).
实施例27:
4-[3-(3-溴-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物27)
Figure PCTCN2016110007-appb-000076
标题化合物采用实施例1方法合成,用3-溴-5,6,7,8-四氢-[1,2,4]三氮唑并[4,3-a]吡嗪取代实施例1中所用的4,5,6,7-四氢-2H-吡唑并[3,4-c]吡啶。
MS m/z(ESI):483[M+H]+
1HNMR(400MHz,CD3OD)δ:8.36(t,1H,J=8Hz),7.99-7.80(m,3H),7.57-7.50(m,1H),7.46-7.41(m,1H),7.24-7.17(m,1H),5.10-5.00(m,1H),4.76-4.59(m,1H),4.42-4.35(m,2H),4.26-3.93(m,4H).
实施例28:
4-[3-(3-溴-6,7,8,9-四氢-5H-[1,2,4]三氮唑并[4,3-d][1,4]二氮杂卓-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物28)
Figure PCTCN2016110007-appb-000077
标题化合物采用实施例1方法合成,用3-溴-6,7,8,9-四氢-5H-[1,2,4]三氮唑并[4,3-d][1,4]二氮杂卓取代实施例1中所用的4,5,6,7-四氢-2H-吡唑并[3,4-c]吡啶。
MS m/z(ESI):497[M+H]+
1HNMR(400MHz,CD3OD)δ:8.40-8.34(m,1H),7.99-7.80(m,3H),7.55-7.49(m,1H),7.43-7.39(m,1H),7.23-7.16(m,1H),4.42-4.33(m,4H),4.10-3.95(m,2H),3.38-3.54(m,2H),3.30-2.25(m,1H),3.04-3.00(m,1H).
实施例29:
4-{4-氟-3-[3-(1-羟基-1-甲基-乙基)-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮(化合物29)
Figure PCTCN2016110007-appb-000078
反应路线:
Figure PCTCN2016110007-appb-000079
第一步:3-(1-羟基-1-甲基-乙基)-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-碳酸叔丁酯
将化合物7-叔丁氧羰基-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-3-碳酸甲酯(200mg,0.71mmol)完全溶解于5mL干燥THF中,氮气保护下室温缓慢加入3mol/L甲基溴化镁(2.37mL,7.1mmol),滴加完毕后转至45℃搅拌至LCMS监测反应完全。降至室温后,加入饱和氯化铵溶液淬灭反应,以二氯甲烷萃取,饱和食盐水返洗,硫酸钠干燥,减压浓缩得到棕色油状物,无需纯化直接用于下一步反应。
第二步:2-(5,6,7,8-四氢-咪唑并[1,2-a]吡嗪-3-基)-丙基-2-醇
将上一步所得化合物完全溶解于10mL二氯甲烷,加入1mL三氟醋酸,转至35℃搅拌至LCMS监测反应完全,减压浓缩除去二氯甲烷与三氟醋酸,所得粗品直接用于下一 步反应。
第三步:4-{4-氟-3-[3-(1-羟基-1-甲基-乙基)-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮
采用实施例1方法合成,用2-(5,6,7,8-四氢-咪唑并[1,2-a]吡嗪-3-基)-丙基-2-醇三氟乙酸盐取代实施例1中所用的4,5,6,7-四氢-2H-吡唑并[3,4-c]吡啶。
MS m/z(ESI):462[M+H]+
1HNMR(400MHz,CD3OD)δ:8.39-8.34(m,1H),7.98-7.94(m,1H),7.92-7.80(m,2H),7.54-7.41(m,2H),7.23-7.16(m,1H),6.83-6.76(m,1H),4.90-4.87(m,1H),4.57-4.54(m,1H),4.43-4.36(m,3H),4.27-4.14(m,2H),3.73-3.68(m,1H),1.57(s,6H).
实施例30:
N-甲基-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-2-三氟甲基-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-3-甲酰胺(化合物30)
Figure PCTCN2016110007-appb-000080
第一步:7-[2-氟-5-(4-氧杂-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-2-三氟甲基-5,6-二氢-8H-咪唑[1,2-a]吡嗪-3-羧酸乙酯
合成方法参见实施例18。
第二步:采用实施例4第二步的方法合成。用第一步的7-[2-氟-5-(4-氧杂-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-2-三氟甲基-5,6-二氢-8H-咪唑[1,2-a]吡嗪-3-羧酸乙酯取代实施例4第二步中所用的7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-2-甲酸甲酯。
MS m/z(ESI):529[M+H]+
1HNMR(400MHz,CD3OD)δ:8.39-8.35(m,1H),7.98-7.94(m,1H),7.91-7.80(m,2H),7.55-7.49(m,1H),7.46-7.43(m,1H),7.23-7.17(m,1H),4.99-4.95(m,1H),4.62-4.59(m,1H),4.42-4.37(m,2H),4.22-3.70(m,4H).
实施例31:
N-乙基-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-2-三氟甲基-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-3-甲酰胺(化合物31)
Figure PCTCN2016110007-appb-000081
第一步:7-[2-氟-5-(4-氧杂-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-2-三氟甲基-5,6-二氢-8H-咪唑[1,2-a]吡嗪-3-羧酸乙酯
合成方法参见实施例18。
第二步:采用实施例4第二步的方法合成。用第一步的7-[2-氟-5-(4-氧杂-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-2-三氟甲基-5,6-二氢-8H-咪唑[1,2-a]吡嗪-3-羧酸乙酯取代实施例4第二步中所用的7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-2-甲酸甲酯。
MS m/z(ESI):543[M+H]+
1HNMR(400MHz,CD3OD)δ:8.40-8.34(m,1H),7.99-7.80(m,3H),7.56-7.42(m,2H),7.23-7.17(m,1H),4.99-4.95(m,1H),4.63-4.58(m,1H),4.40(br s,2H),4.25-3.72(m,4H),3.44-3.35(q,2H,J=7Hz),1.20(t,3H,J=7Hz).
实施例32:
4-{4-氟-3-[3-(1-羟基-1-甲基-乙基)-2-三氟甲基-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮(化合物32)
Figure PCTCN2016110007-appb-000082
反应路线:
Figure PCTCN2016110007-appb-000083
第一步:3-(1-羟基-1-甲基-乙基)-2-三氟甲基-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-碳酸叔丁酯
采用实施例29第一步的方法合成。
第二步:2-(2-三氟甲基-5,6,7,8-四氢-咪唑并[1,2-a]吡嗪-3-基)-丙基-2-醇
采用实施例29第二步的方法合成。
第三步:4-{4-氟-3-[3-(1-羟基-1-甲基-乙基)-2-三氟甲基-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮
采用实施例1的方法合成,用2-(2-三氟甲基-5,6,7,8-四氢-咪唑并[1,2-a]吡嗪-3-基)-丙基-2-醇取代实施例1中所用的4,5,6,7-四氢-2H-吡唑并[3,4-c]吡啶。
MS m/z(ESI):529[M+H]+
1HNMR(400MHz,CD3OD)δ:8.40-8.34(m,1H),7.99-7.79(m,3H),7.56-7.42(m,2H),7.23-7.16(m,1H),4.99-4.95(m,1H),4.63-4.58(m,1H),4.44-4.38(m,2H),4.25-3.72(m,4H),1.30(s,6H).
实施例33:
4-[4-氟-3-(3-甲氧基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-苄基]-2H-酞嗪-1-酮(化合物33)
Figure PCTCN2016110007-appb-000084
反应路线:
Figure PCTCN2016110007-appb-000085
第一步:3-甲氧基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-碳酸叔丁酯
将化合物3-溴-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-碳酸叔丁酯(100mg,0.33mmol)完全溶解于5mLMeOH中,加入甲醇钠(54mg,1mmol)室温搅拌至LCMS监测反应完全。以二氯甲烷萃取,饱和食盐水洗涤,硫酸钠干燥,减压浓缩得到白色固体(84mg,收率99%)。
第二步:3-甲氧基-5,6,7,8-四氢-[1,2,4]三氮唑并[4,3-a]吡嗪
采用实施例32第二步的方法合成。
第三步:4-[4-氟-3-(3-甲氧基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-苄基]-2H-酞嗪-1-酮
采用实施例1的方法合成,用3-甲氧基-5,6,7,8-四氢-[1,2,4]三氮唑[4,3-a]吡嗪取代实施例1中所用的4,5,6,7-四氢-2H-吡唑并[3,4-c]吡啶。
MS m/z(ESI):435[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.59(br s,1H),8.30-8.24(m,1H),7.99-7.80(m, 3H),7.52-7.40(m,2H),7.32-7.24(m,1H),4.95-4.70(m,1H),4.33(s,3H),4.15-3.98(m,4H),3.82-3.55(m,3H).
实施例34:
N-(2-甲氧基乙基)-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺(化合物34)
Figure PCTCN2016110007-appb-000086
第一步:7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酸乙酯
合成方法参见实施例5。
第二步:N-(2-甲氧基乙基)-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺
采用实施例4第二步的方法合成,用第一步的7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酸乙酯取代实施例4中所用的7-[2-氟-5-(4-氧代-3,4-二氢-1-酞嗪基-甲基)-苯甲酰基]-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-2-甲酸甲酯,用甲氧基乙胺取代实施例4中所用的甲胺溶液。
MS m/z(ESI):506[M+H]+
1HNMR(400MHz,DMSO-d6)δ:12.60(br s,1H),8.89-8.82(m,1H),8.29-8.24(m,1H),8.00-7.80(m,3H),7.53-7.45(m,2H),7.31-7.25(m,1H),5.08-4.64(m,2H),4.40-4.04(m,6H),3.50-3.38(m,2H),3.26(s,3H).
实施例35:
N-(2-羟基-2-甲基丙基)-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺(化合物35)
Figure PCTCN2016110007-appb-000087
第一步:7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酸乙酯
合成方法参见实施例5。
第二步:N-(2-羟基-2-甲基丙基)-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺
采用实施例4第二步的方法合成,用第一步的7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酸乙酯取代实施例4中所用的7-[2-氟-5-(4-氧代-3,4-二氢-1-酞嗪基-甲基)-苯甲酰基]-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-2-甲酸甲酯,用1-氨基-2-甲基丙基-2-醇取代实施例4中所用的甲胺溶液。
MS m/z(ESI):520[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.59(br s,1H),8.47-8.38(m,1H),8.28-8.23(m,1H),8.00-7.80(m,3H),7.52-7.45(m,2H),7.31-7.24(m,1H),5.08-4.40(m,1H),4.70-4.24(m,6H),3.67-3.60(m,1H),3.26(s,3H),3.24(d,2H,J=6Hz),1.11(s,6H).
实施例36:
N-(2-甲基磺酰基乙基)-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺(化合物36)
Figure PCTCN2016110007-appb-000088
第一步:7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酸乙酯
合成方法参见实施例5。
第二步:N-(2-甲基磺酰基乙基)-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺
采用实施例4第二步的方法合成,用第一步的7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酸乙酯取代实施例4中所用的7-[2-氟-5-(4-氧代-3,4-二氢-1-酞嗪基-甲基)-苯甲酰基]-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-2-甲酸甲酯,用2-(甲基磺酰基)乙胺取代实施例4中所用的甲胺溶液。
MS m/z(ESI):554[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.60(br s,1H),9.16-9.08(m,1H),8.26(d,1H,J=8Hz),8.00-7.80(m,3H),7.52-7.45(m,2H),7.32-7.25(m,1H),5.08-4.64(m,2H),4.40-4.04(m,4H),3.72-3.35(m,6H),3.04(s,3H).
实施例37:
4-[3-(3-乙酰基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物37)
Figure PCTCN2016110007-appb-000089
反应路线:
Figure PCTCN2016110007-appb-000090
第一步:3-乙酰基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-碳酸叔丁酯
合成方法参见实施例29第一步,用7-叔丁基-3-乙基-5,6-二氢-[1,2,4]三氮唑并[4,3-a]吡嗪-3,7(8H)-二甲酸酯取代实施例29第一步的7-叔丁氧羰基-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-3-甲酸甲酯,并且甲基格式试剂的用量降低为2当量。
第二步:1-(5,6,7,8-四氢-[1,2,4]三氮唑并[4,3-a]吡嗪-3-基)乙酮三氟乙酸盐
将第一步所得化合物3-乙酰基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-碳酸叔丁酯溶于二氯甲烷,然后加入三氟乙酸加热至反应完全。反应液浓缩后所得混合物直接用于下步反应。
第三步:4-[3-(3-乙酰基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮
合成方法参见实施例29第三步,用1-(5,6,7,8-四氢-[1,2,4]三氮唑并[4,3-a]吡嗪-3-基)乙酮三氟乙酸盐取代实施例29第三步中所用的2-(5,6,7,8-四氢-咪唑并[1,2-a]吡嗪-3-基)-丙基-2-醇三氟乙酸盐。
MS m/z(ESI):447[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.60(br s,1H),8.29-8.24(m,2H),7.99-7.81(m,3H),7.52-7.43(m,2H),7.32-7.25(m,1H),5.03-4.70(m,2H),4.39-4.15(m,4H),3.97-3.58(m,2H),2.64(s,3H).
实施例38:
4-{4-氟-3-[3-(1-甲基-1H-吡唑-4-基)-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮(化合物38)
Figure PCTCN2016110007-appb-000091
反应路线:
Figure PCTCN2016110007-appb-000092
第一步:3-(1-甲基-1H-吡唑-4-基)-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-碳酸叔丁酯
将1-甲基-1H-吡唑-4-硼酸(265mg,2.1mmol),3-溴-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-碳酸叔丁酯(604mg,2mmol)溶于DME(5mL),超声波下脱气然后用氮气置换后加入三苯基膦氯化钯(30mg)和2N的碳酸钠水溶液(1mL)。该混合物在85度下反应5小时,反应完全后加入水(3mL)用二氯甲烷萃取(10mLx4)。二氯甲烷层用无水硫酸镁干燥,浓缩后,薄层TLC纯化得目标产物200mg,收率:66%。
第二步:3-(1-甲基-1H-吡唑-4-基)-5,6,7,8-四氢-咪唑并[1,2-a]吡嗪盐酸盐
将第一步所得产物溶于2mL甲醇,然后加入氯化氢的1,4-二氧六环饱和溶液(2mL,4N)室温反应至反应完全,减压蒸除溶剂,所得残余物直接用于下一步反应。
第三步:4-{4-氟-3-[3-(1-甲基-1H-吡唑-4-基)-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮
合成参见实施例29第三步,用3-(1-甲基-1H-吡唑-4-基)-5,6,7,8-四氢-咪唑并[1,2-a]吡嗪盐酸盐取代实施例29中的2-(5,6,7,8-四氢-咪唑并[1,2-a]吡嗪-3-基)-丙基-2-醇三氟乙酸盐。
MS m/z(ESI):485[M+H]+
1H NMR(400MHz,CD3OD)δ:8.36-8.30(m,1H),7.96-7.77(m,4H),7.68-7.59(m,1H),7.55-7.41(m,2H),7.24-7.14(m,1H),7.03-6.95(m,1H),4.96-4.90(m,1H),4.63-4.35(m,3H),4.27-4.09(m,2H),3.99-3.88(m,4H),3.77-3.70(m,1H)。
实施例39:
4-{4-氟-3-[3-(1-甲基-1H-吡唑-4-基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基]- 苄基}-2H-酞嗪-1-酮(化合物39)
Figure PCTCN2016110007-appb-000093
反应路线:
Figure PCTCN2016110007-appb-000094
第一步:3-(1-甲基-1H-吡唑-4-基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-碳酸叔丁酯
合成方法参见实施例38第一步用3-溴-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-碳酸叔丁酯取代实施例38中的3-溴-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-碳酸叔丁酯。
第二步:3-(1-甲基-1H-吡唑-4-基)-5,6,7,8-四氢-[1,2,4]三氮唑并[4,3-a]吡嗪盐酸盐
合成参见实施例38第二步,用3-(1-甲基-1H-吡唑-4-基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-碳酸叔丁酯取代实施例38中的3-(1-甲基-1H-吡唑-4-基)-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-碳酸叔丁酯。
第三步4-{4-氟-3-[3-(1-甲基-1H-吡唑-4-基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮
合成参见实施例29第三步,用3-(1-甲基-1H-吡唑-4-基)-5,6,7,8-四氢-[1,2,4]三氮唑并[4,3-a]吡嗪盐酸盐取代实施例29中的2-(5,6,7,8-四氢-咪唑并[1,2-a]吡嗪-3-基)-丙基-2-醇三氟乙酸盐。
MS m/z(ESI):486[M+H]+
1H NMR(400MHz,CD3OD)δ:8.37-8.30(m,1H),8.20-8.10(m,1H),7.97-7.77(m,4H),7.58-7.44(m,2H),7.25-7.17(m,1H),5.13-5.04(m,1H),4.95-4.90(m,1H),4.79-4.70(m,1H),4.41-4.36(m,2H),4.32-4.21(m,1H),4.12-4.04(m,1H),4.00-3.97(m,3H),3.83-3.74(m,1H)。
实施例40:
4-[4-氟-3-(3-甲巯基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-苄基]-2H-酞嗪-1-酮(化合物40)
Figure PCTCN2016110007-appb-000095
反应路线:
Figure PCTCN2016110007-appb-000096
第一步:3-甲巯基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-碳酸叔丁酯
合成方法参见实施例33第一步,用甲硫醇钠取代实施例33第一步中用的甲醇钠,溶剂改为DMF。
第二步:3-甲巯基-5,6,7,8-四氢-[1,2,4]三氮唑并[4,3-a]吡嗪三氟乙酸盐
合成方法参见实施例33第二步。
第三步:4-[4-氟-3-(3-甲巯基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-苄基]-2H-酞嗪-1-酮
合成方法参见实施例33第三步。
MS m/z(ESI):451[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.59(br s,1H),8.29-8.24(m,1H),7.99-7.80(m,3H),7.52-7.43(m,2H),7.34-7.23(m,1H),4.60-4.39(m,3H),3.96-3.91(m,1H),3.75-3.58(m,3H),3.19-3.15(m,1H),2.61-2.56(s,3H)。
实施例41:
4-[4-氟-3-(3-甲基磺酰基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-苄基]-2H-酞嗪-1-酮(化合物41)
Figure PCTCN2016110007-appb-000097
Figure PCTCN2016110007-appb-000098
反应路线:
Figure PCTCN2016110007-appb-000099
第一步:3-甲巯基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-碳酸叔丁酯
合成方法参见实施例33第一步,用甲硫醇钠取代实施例33第一步中用的甲醇钠,溶剂改为DMF。
第二步:3-甲基磺酰基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-碳酸叔丁酯
0℃下,向10mL3-甲巯基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-碳酸叔丁酯(270mg,1mmol)的二氯甲烷溶液中加入间氯过氧苯甲酸(519mg,3mmol),自然升至室温,TLC跟踪监测,5小时后原料反应完全。向反应液中加入10%亚硫酸钠溶液,搅拌30分钟后再加入10%碳酸钠溶液,继续搅拌10分钟,用二氯甲烷(50mL×4)萃取,合并有机相,无水硫酸钠干燥,旋干,再用甲基叔丁基醚洗涤后得到目标分子,收率:85%)。
第三步:3-甲基磺酰基-5,6,7,8-四氢-[1,2,4]三氮唑并[4,3-a]吡嗪三氟乙酸盐
合成方法参见实施例33第二步。
第四步:4-[4-氟-3-(3-甲基磺酰基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-苄基]-2H-酞嗪-1-酮
合成方法参见实施例33第三步。
MS m/z(ESI):483[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.60(br s,1H),8.28-8.24(m,1H),7.98-7.80(m,3H),7.55-7.42(m,2H),7.34-7.26(m,1H),4.75-4.69(m,1H),4.31(s,3H),4.20-4.07(m,2H),3.73-3.45(m,5H)。
实施例42:
4-{4-氟-3-[3-(2-羟基丙基-2-基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮(化合物42)
Figure PCTCN2016110007-appb-000100
Figure PCTCN2016110007-appb-000101
反应路线:
Figure PCTCN2016110007-appb-000102
第一步:3-(2-羟基丙基-2-基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-碳酸叔丁酯
合成方法参见实施例29第一步,用7-叔丁氧羰基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酸乙酯取代实施例29中的7-叔丁氧羰基-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-3-甲酸甲酯。
第二步:2-(5,6,7,8-四氢-[1,2,4]三氮唑并[4,3-a]吡嗪-3-基)-丙基-2-醇三氟乙酸盐
合成方法参见实施例29第二步,用3-(2-羟基丙基-2-基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-碳酸叔丁酯取代实施例29中的3-(1-羟基-1-甲基-乙基)-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-碳酸叔丁酯。
第三步:4-{4-氟-3-[3-(2-羟基丙基-2-基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮
合成方法参见实施例29第三步,用2-(5,6,7,8-四氢-[1,2,4]三氮唑并[4,3-a]吡嗪-3-基)-丙基-2-醇三氟乙酸盐取代实施例29中的2-(5,6,7,8-四氢-咪唑并[1,2-a]吡嗪-3-基)-丙基-2-醇三氟乙酸盐。
MS m/z(ESI):463[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.60(br s,1H),8.28-8.24(m,1H),7.99-7.81(m,3H),7.53-7.43(m,2H),7.33-7.24(m,1H),4.95-4.54(m,2H),4.38-4.24(m,3H),4.16-4.00(m,2H),3.66-3.56(m,1H),1.52(s,6H).
实施例43:
4-{4-氟-3-[3-(2-羟基丙基-2-基)-1,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基]-苄基}-2H-酞嗪-1-酮(化合物43)
Figure PCTCN2016110007-appb-000103
Figure PCTCN2016110007-appb-000104
反应路线:
Figure PCTCN2016110007-appb-000105
第一步:4,5,6,7-四氢-1H-吡唑并[3,4-c]吡啶-3-甲酸甲酯盐酸盐
将6-叔丁氧羰基-4,5,6,7-四氢-1H-吡唑并[3,4-c]吡啶-3-羧酸(267mg,1mmol)溶于5mL甲醇,然后缓慢加入1mL氯化亚砜。所得溶液回流过夜,LCMS监测反应完全后将甲醇除去,残余物直接用于下一步反应。
第二步:6-叔丁氧羰基-1,4,5,7-四氢-吡唑并[3,4-c]吡啶-3-甲酸甲酯
向第一步所得残余物(250mg)依次加入二氯甲烷(5mL),三乙胺(404mg,4mmol)和二碳酸二叔丁酯(516mg,2mmol)室温反应过夜。然后加入水分液后将二氯甲烷层浓缩,薄层TLC纯化得到目标产物(220mg,收率78.3%)。
第三步:3-(2-羟基丙基-2-基)-1,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-碳酸叔丁酯
合成参见实施例29第一步,用6-叔丁氧羰基-1,4,5,7-四氢-吡唑并[3,4-c]吡啶-3-甲酸甲酯取代实施例29中的7-叔丁氧羰基-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-3-甲酸甲酯。
第四步:2-(4,5,6,7-四氢-1H-吡唑并[3,4-c]吡啶-3-基)丙基-2-醇三氟乙酸盐
合成参见实施例29第二步,用3-(2-羟基丙基-2-基)-1,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-碳酸叔丁酯取代实施例29中的3-(1-羟基-1-甲基-乙基)-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-碳酸叔丁酯。
第五步:4-{4-氟-3-[3-(2-羟基丙基-2-基)-1,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基]-苄基}-2H-酞嗪-1-酮
合成参见实施例29第三步,用2-(4,5,6,7-四氢-1H-吡唑并[3,4-c]吡啶-3-基)丙基-2-醇三氟乙酸盐取代实施例29中的2-(5,6,7,8-四氢-咪唑并[1,2-a]吡嗪-3-基)-丙基-2-醇三氟乙酸盐。
MS m/z(ESI):462[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.62(br s,1H),8.27-8.24(m,1H),8.00-7.96(m,1H),7.93-7.80(m,2H),7.49-7.20(m,3H),5.25-4.94(m,1H),4.71-4.63(m,1H),4.37-4.26(m,3H),4.01-3.73(m,1H),3.72-3.66(m,1H),3.53-3.39(m,1H),3.04(m,6H).
实施例44:
4-{4-氟-3-[3-(1-羟基乙基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮(化合物44)
Figure PCTCN2016110007-appb-000106
反应路线:
Figure PCTCN2016110007-appb-000107
第一步:3-(1-羟基乙基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-碳酸叔丁酯
将3-乙酰基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-碳酸叔丁酯(266mg,1mmol)溶于甲醇(5mL)冰浴下加入氰基硼氢化钠(315mg,5mmol)自然升温至室温,搅拌过夜。将反应液浓缩后,加入水和二氯甲烷,二氯甲烷层用无水硫酸镁干燥,浓缩后得目标产物(250mg,收率93%)。
第二步:1-(5,6,7,8-四氢-[1,2,4]三氮唑并[4,3-a]吡嗪-3-基)-乙醇三氟乙酸盐
合成参见实施例29第二步,用3-(1-羟基乙基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-碳酸叔丁酯取代实施例29中的3-(1-羟基-1-甲基-乙基)-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-碳酸叔丁酯。
第三步:4-{4-氟-3-[3-(1-羟基乙基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮
合成参见实施例29第三步,用1-(5,6,7,8-四氢-[1,2,4]三氮唑并[4,3-a]吡嗪-3-基)乙醇三氟乙酸盐取代实施例29中的2-(5,6,7,8-四氢-咪唑并[1,2-a]吡嗪-3-基)-丙基-2-醇三氟乙酸盐。
MS m/z(ESI):449[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.60(br s,1H),8.30-8.23(m,1H),8.01-7.79(m,3H),7.53-7.40(m,2H),7.33-7.24(m,1H),5.65-5.54(m,1H),5.01-4.79(m,2H),4.34(s,2H),4.22-4.09(m,1H),4.05-3.92(m,2H),3.71-3.55(m,1H),1.50(d,3H,J=4Hz)。
实施例45:
4-{4-氟-[(3-(1-甲基-1H-吡唑-3-基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮(化合物45)
Figure PCTCN2016110007-appb-000108
反应路线:
Figure PCTCN2016110007-appb-000109
第一步:1-甲基-1H-吡唑-3-硼酸
将3-溴-1-甲基-1H-吡唑(322mg,2mmol),联硼酸频哪醇酯(1.016g,4mmol),溶于1,4-二氧六环(10mL),然后加入醋酸钾(392mg,4mmol),所得溶液在超声波下脱气,进行氮气置换后加入三苯基膦氯化钯(32mg)。110度下反应5小时后,减压蒸干溶剂,加入水(10mL)然后用二氯甲烷萃取(10mL×4),二氯甲烷层用无水硫酸镁干燥后,旋干。残余物经薄层TLC纯化得到目标产物179mg,收率71%。
第二步:3-(1-甲基-1H-吡唑-3-基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-碳酸叔丁酯
合成参见实施例38第一步用1-甲基-1H-吡唑-3-硼酸取代实施例38中的1-甲基-1H-吡唑-4-硼酸。
第三步3-(1-甲基-1H-吡唑-3-基)-5,6,7,8-四氢-[1,2,4]三氮唑并[4,3-a]吡嗪盐酸盐
合成参见实施例38第二步,用3-(1-甲基-1H-吡唑-3-基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-碳酸叔丁酯取代实施例38中的3-(1-甲基-1H-吡唑-4-基)-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-碳酸叔丁酯。
第四步:4{4-氟-3-[3-(1-甲基-1H-吡唑-3-基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡 嗪-7-羰基]-苄基}-2H-酞嗪-1-酮
合成参见实施例29第三步,用3-(1-甲基-1H-吡唑-3-基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪盐酸盐取代实施例29中的2-(5,6,7,8-四氢-咪唑并[1,2-a]吡嗪-3-基)-丙基-2-醇三氟乙酸盐。
MS m/z(ESI):485[M+H]+
1H NMR(400MHz,CD3OD)δ:8.42-8.30(m,1H),8.11-7.47(m,7H),7.31-7.20(m,1H),5.16-4.72(m,2H),4.49-4.35(m,4H),4.11-3.91(m,2H),3.51-3.40(m,3H)。
实施例46:
4-{3-[3-(1,1-二氟乙基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基]-4-氟苄基}-2H-酞嗪-1-酮(化合物46)
Figure PCTCN2016110007-appb-000110
反应路线:
Figure PCTCN2016110007-appb-000111
第一步:3-(1,1-二氟乙基)-[1,2,4]三氮唑并[4,3-a]吡嗪
将化合物2-肼基吡嗪(1g,1eq)、2,2-二氟丙酸(3g,3eq)加入圆底烧瓶中,再将PPA将入其中,搅拌均匀后在180℃下反应18小时。LCMS监测,至反应结束。将反应物倒入饱和碳酸氢钠水溶液中,加入二氯甲烷萃取,有机层用无水硫酸钠干燥。蒸除有机溶剂,残余物柱层析纯化得3-(1,1-二氟乙基)-[1,2,4]三氮唑并[4,3-a]吡嗪450mg,收率27%。
第二步:3-(1,1-二氟乙基)-5,6,7,8-[1,2,4]三氮唑并[4,3-a]吡嗪
将化合物3-(1,1-二氟乙基)-[1,2,4]三氮唑并[4,3-a]吡嗪(450mg)溶于乙醇中,再向其中加入Pd/C(50mg)。在氢气氛下反应过夜。LCMS监测,至反应结束蒸干乙醇得3-(1,1-二氟乙基)-5,6,7,8-[1,2,4]三氮唑并[4,3-a]吡嗪462mg。
第三步:4-{3-[3-(1,1-二氟乙基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基]-4- 氟苄基}-2H-酞嗪-1-酮(化合物46)
将化合物3-(1,1-二氟乙基)-5,6,7,8-[1,2,4]三氮唑并[4,3-a]吡嗪(462mg,1eq),2-氟-5-((4-氧代-3,4-二氢酞嗪-1-基)甲基)苯甲酸(806mg,1.1eq)溶于DMF中,再将T3P(1.2eq),DIEA(1.27g,4eq)加入其中,室温搅拌,LCMS检测至反应结束。将反应物倾入饱和碳酸氢钠水溶液中加入二氯甲烷萃取,有机层用无水硫酸钠干燥。蒸干有机溶剂,残余物经柱层析纯化得目标化合物800mg,收率69.6%。
MS m/z(ESI):469[M+H]+
1HNMR(400MHz,DMSO-d6)δ:12.59(s,1H),8.29-8.22(m,1H),7.98-7.80(m,3H),7.54-7.45(m,2H),7.35-7.24(m,1H),5.10-4.61(m,2H),4.34(s,2H),4.27-3.96(m,3H),3.71-3.62(m,1H),2.16(t,3H,J=16Hz).
实施例47:
4-[4-氟-3-(3-五氟乙基-5,6,7,8-四氢-[1,2,4]三唑[4,3-a]吡嗪-7-羰基)苄基]-2H-酞嗪-1-酮(化合物63)
Figure PCTCN2016110007-appb-000112
反应路线:
Figure PCTCN2016110007-appb-000113
第一步:3-五氟乙基-[1,2,4]三唑[4,3-a]吡嗪
室温下向吡嗪-2-肼(10.0g,90.8mmol)中加入80mL PPA,五氟丙酸(37.2g,227.0mmol),升温至110℃反应至吡嗪-2-肼反应完全。搅拌下,将反应液缓慢倾倒入1L冰水中,碳酸氢钠调节pH呈弱碱性,用乙酸乙酯(150mL×4)萃取,合并有机相,饱和食盐水洗涤,无水硫酸钠干燥,浓缩后得到的残余物经柱层析(PE/EA=4/1)纯化后得到目标产物(17.8g,收率:82.4%)。
MS m/z(ESI):239.1[M+H]+.
第二步:3-五氟乙基-5,6,7,8-四氢-[1,2,4]三唑[4,3-a]吡嗪
室温下,向3-五氟乙基-[1,2,4]三唑[4,3-a]吡嗪(15.0g,63.0mmol)中加入150ml无水乙醇,搅拌澄清后加入2.0g 10%Pd/C,氢气置换后于室温下反应至原料完全消失。硅藻土垫滤,并以100ml无水乙醇洗涤滤饼,减压浓缩得目标产物(13.6g,收率:88.9%)未经纯化,直接用于下一步。
MS m/z(ESI):243.1[M+H]+.
第三步:4-[4-氟-3-(3-五氟乙基-5,6,7,8-四氢-[1,2,4]三唑[4,3-a]吡嗪-7-羰基)苄基]-2H-酞嗪-1-酮
室温下,将5-(3,4-二氢-4-氧代-1-酞嗪基)甲基-2-氟苯甲酸(1.0g,3.35mmol),HATU(1.40g,3.69mmol)和DIPEA(649.4mg,5.03mmol)混溶于10mLDMF,室温搅拌30min后,加入第二步的3-五氟乙基-5,6,7,8-四氢-[1,2,4]三唑[4,3-a]吡嗪(0.89g,3.69mmol),室温反应至5-(3,4-二氢-4-氧代-1-酞嗪基)甲基-2-氟苯甲酸完全消失后,向反应液中加入150mL水,用乙酸乙酯(30mL×4)萃取,合并有机相,饱和食盐水洗涤,无水硫酸钠干燥,浓缩后得到的残余物经柱层析(DCM/Acetone=3/1)纯化后得到目标产物(1.18g,收率:67.4%)。
MS m/z(ESI):523.2[M+H]+.
1HNMR(400MHz,DMSO-d6)δ:12.60(s,1H),8.27-8.24(m,1H),7.97-7.81(m,3H),7.53-7.44(m,2H),7.33-7.27(m,1H),5.04-4.95(m,1H),4.72-4.65(m,1H),4.35(s,2H),4.29-4.15(m,2H),4.02-3.98(m,1H),3.68-3.62(m,1H).
参照实施例中所述合成方法,本申请还合成了以下化合物:
表1
Figure PCTCN2016110007-appb-000114
Figure PCTCN2016110007-appb-000115
Figure PCTCN2016110007-appb-000116
Figure PCTCN2016110007-appb-000117
Figure PCTCN2016110007-appb-000118
Figure PCTCN2016110007-appb-000119
生物学活性
实验例1PARP1激酶活性筛选
PARP-1抑制剂的酶活筛选采用BPS Bioscience的PARP1化学发光试剂盒进行。
试剂盒:PARP1Chemiluminescent Assay Kit;
厂家:BPS Bioscience;
实验方法
酶标板预处理:从包装中取出微孔板,然后加入50μL/孔的1×PARP缓冲稀释的1×组蛋白,4℃孵育过夜;次日弃去微孔板中的液体,并采用200μL/孔PBST(1×PBS及1%Triton X-100)洗板,除去全部清洗液;然后每孔加入200μL的终止缓冲液,室温孵育90min后弃去液体;并采用200μL/孔PBST洗板,最后除去全部清洗液。
反应:按下表所列步骤与量依次加入底物和酶及测试化合物等,室温孵育1h。
表2反应体系
Figure PCTCN2016110007-appb-000120
Figure PCTCN2016110007-appb-000121
检测:反应结束后向每孔中加入50μL稀释后的Strep-HRP,室温培养;用PBST洗孔板后将清洗溶液除去;混合HRP底物A和B,然后加入各孔100μL;最后化学发光法(Luminometric Measurement)检测待测化合物对PARP1激酶抑制活性。
结果:
表3已测试化合物单点浓度抑制率
化合物ID 单点浓度抑制率%
奥拉帕尼 77.47%(5nM)
化合物1 88.77%(5nM)
化合物5 92.78%(5nM)
化合物13 99.70%(5nM)
化合物14 99.46%(5nM)
化合物15 99.50%(5nM)
化合物16 96.65%(5nM)
化合物17 97.88%(5nM)
化合物18 86.10%(5nM)
化合物19 90.90%(5nM)
化合物20 99.65%(5nM)
化合物21 98.97%(5nM)
化合物22 86.27%(5nM)
化合物23 78.15%(5nM)
化合物24 99.48%(5nM)
化合物25 98.13%(5nM)
化合物26 89.37%(5nM)
化合物27 92.66%(5nM)
化合物28 78.07%(5nM)
化合物29 81.90%(5nM)
化合物33 88.84%(5nM)
化合物34 88.12%(5nM)
化合物35 83.08%(5nM)
由表3可以看出,本申请的化合物对PARP1激酶活性具有明显的抑制作用,并且化 合物抑制活性显著优于阳性药物。
实验例2细胞增殖实验
采用CCK-8法测定化合物对细胞增殖的影响。
试剂盒:CCK-8试剂盒(Cell Counting Kit-8)
厂家:碧云天(Beyotime)
实验方法
细胞培养:根据以下条件培养细胞,细胞细化后采用细胞计数器计数,并根据以下要求调整细胞至所需浓度,然后每孔接种细胞100μL,接种后24小时给药。
表4细胞培养条件及种板密度
细胞名称 培养液 培养箱条件
乳腺癌MDA-MB-468 DMEM/F12+10%FBS 5%CO2,37℃
结直肠癌HCT116 1640+10%FBS 5%CO2,37℃
胰腺癌Capan-1 IMEM+20%FBS 5%CO2,37℃
化合物配制:培养一天后,用DMSO溶解实验药品和奥拉帕尼制成母液,吸取适量母液至培养液中混匀,药品溶液配置为相应的孵育浓度。
孵育时间:给药后将于培养箱中继续孵育7天。
检测:孵育结束后换新鲜培养液200μL/孔置于培养基中稳定2h后加20μL/孔的CCK8孵育3h以上,在450nm波长,参考波长650nm进行双波长测定吸光度。IC50值用GraphPad计算得到。
结果详见表5-1,5-2,5-3:
表5-1:对乳腺癌MDA-MB-468的抑制作用
Figure PCTCN2016110007-appb-000122
Figure PCTCN2016110007-appb-000123
表5-1中化合物对乳腺癌MDA-MB-468肿瘤的抑制作用均优于奥拉帕尼。本申请的化合物基本上具有与上述化合物对乳腺癌MDA-MB-468肿瘤类似的抑制作用。本申请的化合物对乳腺癌MDA-MB-436、MDA-MB-453肿瘤具有类似的抑制作用。
表5-2:对胰腺癌Capan-1的抑制作用
Figure PCTCN2016110007-appb-000124
Figure PCTCN2016110007-appb-000125
由表5-2中化合物对胰腺癌Capan-1肿瘤的抑制作用均优于奥拉帕尼。本申请的化合物基本上具有与上述化合物对胰腺癌Capan-1肿瘤类似的抑制作用。
表5-3:对结直肠癌HCT116的抑制作用
Figure PCTCN2016110007-appb-000126
由表5-3中化合物对直肠癌HCT116肿瘤的抑制作用均优于奥拉帕尼。本申请的化合物基本上具有与上述化合物对直肠癌HCT116肿瘤类似的抑制作用。
综上,本申请的化合物对于不同乳腺癌、结直肠癌和胰腺癌等癌细胞均具有优异的抑制活性,表现出良好的抗肿瘤作用。
实验例3化合物的药代动力学(PK)研究
3.1大鼠PK研究
分别通过静脉和灌胃给予雄性SD大鼠本申请化合物和奥拉帕尼,考察药代动力学特点。IV和PO的给药剂量分别是1和5mg/kg,溶媒系统均为10%DMSO:10%solutol:80%生理盐水。IV和PO给药后在不同时间点收集血液,血液采用肝素钠抗凝,离心后得到血浆样品保存于-80℃。血浆样品经已经沉淀蛋白处理后进行LC-MS/MS分析。
LC-MS/MS,色谱柱为Waters X-Bridge C18柱(21mm*50mm,3.5μm);流动相A相为水+2mM乙酸铵,B相为甲醇+2mM乙酸铵,流速为0.4mL/min,柱温为40℃。采用离子源为ESI源正离子模式,扫描方式为多重反应监测(MRM)。
应用WinNonlin 6.3软件,采用非房室模型计算药代动力学参数,结果见表6-1,6-2.
表6-1:大鼠体内的IV给药药代动力学参数
Figure PCTCN2016110007-appb-000127
根据IV给药方式的结果,本申请化合物在大鼠体内的药代动力学参数均优于奥拉帕尼。
表6-2:大鼠体内的PO给药药代动力学参数
Figure PCTCN2016110007-appb-000128
根据PO给药方式的结果,本申请化合物在大鼠体内的药代动力学参数均优于奥拉帕尼。
另外,在本药代动力学试验中意外发现,奥拉帕尼的半衰期是1.31小时,本申请化合物1的半衰期为2.56小时,化合物5的半衰期为2.52小时,化合物物46的半衰期为2.8小时,由此表明本申请化合物2、化合物5、化合物46的药效作用时间更长久。本申请的其他化合物基本上具有类似的药效性质。
实验例4安全性试验
4.1、hERG测试
在心肌细胞中,human Ether-a-go-go Related Gene(hERG)编码的钾通道介导一种延迟整流钾电流(IKr)。IKr抑制是药物导致QT间期延长最重要的机制。在hERG测试中,判定标准为若待测化合物IC50>30μM,则判定待测化合物对hERG无抑制作用。
采用PredictorTM hERG Fluorescence Polarization Assay,检测本申请化合物对hERG钾离子通道的作用,测试浓度为3,10,30μM。
试验中显示本申请化合物对于hERG的50%抑制浓度(IC50)值均大于30μM。因 此,对hERG均无抑制作用,表明本申请化合物不会导致心脏QT间期延长的安全性隐患。
4.2小鼠急毒试验
通过灌胃给予KM小鼠,考察待测化合物单次给药毒性反应,初步判断最大耐受量(MTD)。
剂量设置为,化合物1:300mg/kg;化合物2、3、6:200、300mg/kg,单次灌胃给药,溶媒为10%DMSO加50%PEG,给药后观察7天。
单次给药高剂量下的耐受性本申请的化合物均有良好的耐受性。
实施例5体内抑瘤实验
本实施例用于评价本申请化合物不同给药途径对肿瘤增殖的抑制有效性。
本实施例考察人胰腺癌细胞株Capan-1皮下移植瘤小鼠通过PO给药途径施用本申请化合物样品后,测试各样品对胰腺癌Capan-1荷瘤鼠的药效。
选择肿瘤体积100-200mm3的荷瘤鼠随机分为8组,7只/组,给药体积为10ml/kg,每天1次给药,共约两周。给药后一周2次测量肿瘤体积。肿瘤体积的计算公式为:V=0.5a×b2,a和b分别表示肿瘤的长径和短径。待测化合物的抑瘤疗效用肿瘤生长抑制率TGI(%)评价。
TGI(%)=[1-(VT末-VT始)/(VC末-VC始)]*100%
其中VT末:处理组实验结束时肿瘤体积均值
VT始:处理组给药开始时肿瘤体积均值
VC末:溶媒对照组实验结束时肿瘤体积均值
VC始:溶媒对照组给药开始时肿瘤体积均值
本试验条件下,本申请的所述化合物具有优良的药效,在10ml/kg等给药剂量下肿瘤部分消退,药效均优于阳性对照组。
制剂例1片剂
速释片剂的组成由表10所示:
表10速释片剂的组成
成分 mg/片剂 占片剂芯重量的百分比(%)
化合物1 100.00 25.00
乳糖 238.00 50.00
微晶纤维素 40.00 10.00
交联羧甲基纤维素钠 16.00 4.00
月桂基硫酸钠 2.00 0.50
硬脂酸镁 4.00 1.00
片剂芯重量 400.00 /
制备方法
使用直接压制法制造标准速释片剂。将化合物1和乳糖、微晶纤维素、交联羧甲基纤维素钠和月桂基硫酸钠称入玻璃小管至占据该小管体积的大约75%,随后在转鼓混合机中一起混合30分钟。该掺合材料经40目(425μm)筛子筛分,随后再转鼓混合15分钟。随后加入硬脂酸镁并用手摇振该掺合物大约20秒。随后将所得混合物分配成400毫克的小份,并使用配有10毫米模具的手压机用0.5吨目标压缩力压制成片剂芯。
制剂例2胶囊制剂
胶囊制剂的组成由表11
表11胶囊制剂的组成
Figure PCTCN2016110007-appb-000129
制备方法,步骤如下:
(1)将上述质量份的化合物5、乳糖、微晶纤维素、微粉硅胶、硬脂酸镁过80目筛,85-90℃干燥约4小时;
(2)将乳糖、微晶纤维素和微粉硅胶混合均匀,然后将化合物5与其混合,进行干法制粒,所得的颗粒过100目筛,并收集20-60目的干颗粒;
(3)将20目以上物料进行再粉碎后,与60目以下物料进行二次干法制粒,将颗粒中加入上述质量份的硬脂酸镁,混合均匀得混合粒,将混合粒置于胶囊填充机上制成胶囊。
尽管本申请的具体实施方式已经得到详细的描述,本领域技术人员将会理解,根据已经公开的所有教导,可以对那些细节进行各种修改和替换,这些改变均在本申请的保护范围之内。本申请的全部范围由所附权利要求及其任何等同物给出。

Claims (29)

  1. 式I化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型,
    Figure PCTCN2016110007-appb-100001
    其中,
    A和B与相连的原子一起形成脂环、脂杂环、芳环或杂芳环;任选地,其中所述脂环、脂杂环、芳环或杂芳环各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、羧基、硝基、烷基、卤代烷基、烯基、炔基、环烷基、脂杂环基、芳基、杂芳基、RO-、ROC(O)-、RC(O)O-、RC(O)-、RR’N-、RC(O)N(R”)-、RR’NC(O)-和RS(O)a-的取代基取代,其中a为0、1或2;
    D和E各自独立地选自C和N,并与相连的原子一起形成5-10元环X,其中所述环X为脂环、脂杂环、芳环或杂芳环;优选地,所述环X为含氮脂杂环或杂芳环;优选地,所述环X为五元或六元含氮脂杂环或杂芳环;优选地,所述环X为五元或六元含氮杂芳环;优选地,所述环X例如为吡咯环、咪唑环、吡唑环、三氮唑环、吡啶环或哌啶酮环;
    m和n各自独立地选自0、1、2和3,且m+n=3或4;
    R1、R2、R3和R4各自独立地选自氢、卤素、羟基、氨基、氰基、羧基、硝基、烷基、烯基、炔基、环烷基、脂杂环基、芳基、杂芳基、RO-、ROC(O)-、RC(O)O-、RC(O)-、RR’N-、RS(O)a-、RC(O)N(R”)-和RR’NC(O)-;任选地,其中所述烷基、烯基、炔基、环烷基、脂杂环基、芳基、杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、羧基、硝基、C1-4烷基、C1-4烷氧基、3-8元环烷基、5-8元脂杂环基、6-10元芳基、5-10元杂芳基和RSO2-的取代基取代,其中a为0、1或2;
    R5选自氢、氧基、卤素、羟基、氨基、氰基、羧基、硝基、烷基、烯基、炔基、环烷基、脂杂环基、芳基、杂芳基、RO-、RC(O)-、ROC(O)-、RC(O)O-、RR’N-、RC(O)N(R”)-、RR’NC(O)-、ROC(O)N(R’)-、RR’NC(O)N(R”)-、RS(O)a-、RR’NSO2-和RS(O)2N(R’)-;任选地,其中所述烷基、烯基、炔基、环烷基、脂杂环基、芳基、杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、羧基、硝基、C1-4烷基、C1-4烷氧基、3-8元环烷基、5-8元脂杂环基、6-10元芳基、5-10元杂芳基和RSO2-的取代基取代,其中a为0、1或2;或者,
    R5并在与之相连的环X上形成多环系统,其中,R5选自脂环、脂杂环、芳环和杂芳环;
    R6不存在或选自氢、氧基、卤素、羟基、氨基、氰基、羧基、硝基、烷基、烯基、炔基、环烷基、脂杂环基、芳基、杂芳基、RO-、RC(O)-、ROC(O)-、RC(O)O-、RR’N-、RC(O)N(R”)-、RR’NC(O)-、ROC(O)N(R’)-、RR’NC(O)N(R”)-、RS(O)a-、RR’NSO2-和RS(O)2N(R’)-;任选地,其中所述烷基、烯基、炔基、环烷基、脂杂环基、芳基、杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、羧基、硝基、C1-4烷基、C1-4烷氧基、3-8元环烷基、5-8元脂杂环基、6-10元芳基、5-10元杂芳基和RSO2-的取代基取代,其中a为0、1或2;
    R、R’和R”各自独立地选自氢、C1-10烷基、3-20元环烷基、5-20元脂杂环基、6-20元芳基和5-20元杂芳基,任选地,其中所述C1-10烷基、3-20元环烷基、5-20元脂杂环基、6-20元芳基或5-20元杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、羧基、硝基、C1-4烷基、卤代C1-4烷基、羟基C1-4烷基、C1-4烷氧基、3-8元环烷基、5-8元脂杂环基、6-10元芳基、5-10元杂芳基和RSO2-的取代基取代,
    其中,所述式I化合物的限制性条件为:
    当A和B与相连的碳原子形成未取代的苯环,R1、R2和R3均为H,R4为F时,
    1)且当式I中
    Figure PCTCN2016110007-appb-100002
    时,R6不存在,R5不为H、羟基、甲基、异丙基、环丙基、三氟甲基、二氟甲基和三氟乙基;
    2)且当式I中
    Figure PCTCN2016110007-appb-100004
    Figure PCTCN2016110007-appb-100005
    时,R5和R6不全为H、且当R5或R6其中一个为H时,另一个不为三氟甲基和乙氧羰基;
    3)式I中
    Figure PCTCN2016110007-appb-100006
    不为
    Figure PCTCN2016110007-appb-100007
    4)所述环X不为吡咯烷、2-吡咯烷酮、吗啉、3-吗啉酮、四氢呋喃、2-噁唑烷酮、四氢噻吩和噻吩烷砜;并且,
    5)所述式I化合物不为4-{[3-(3,4-二氢-1H-吡咯并[1,2-a]吡嗪-2-羰基)-4-氟-苯基]甲基}-2H-酞嗪-1-酮、4-{[3-(6,8-二氢-5H-[1,2,4]三唑并[1,5-a]吡嗪-7-羰基)-4-氟-苯基]甲基}-2H-酞嗪-1-酮、4-{[4-氟-3-(2-三氟甲基-6,8-二氢-5H-[1,2,4]三唑并[1,5-a]吡嗪-7-羰基)-苯基]甲基}-2H-酞嗪-1-酮、和4-[4-氟-3-(5,6-二氢-8H-咪唑并[1,5-a]吡嗪-7-羰基)苄基]-2H-酞嗪-1-酮。
  2. 权利要求1的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型,所述化合物具有式I-A所示结构,
    Figure PCTCN2016110007-appb-100008
    其中,所述式I-A化合物的限制性条件为:所述环X不为吡咯烷、2-吡咯烷酮、吗啉、3-吗啉酮、四氢呋喃、2-噁唑烷酮、四氢噻吩和噻吩烷砜;其余各原子或取代基定义如权利要求1所述。
  3. 权利要求2的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型,其中,所述式I-A化合物中环X为五元杂芳环且杂原子为氮、或六元含氮脂杂环;优选地,所述
    Figure PCTCN2016110007-appb-100009
    选自如下结构:
    Figure PCTCN2016110007-appb-100010
  4. 权利要求2的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型,其中,所述式I-A化合物中所述多环系统选自如下结构:
    Figure PCTCN2016110007-appb-100011
  5. 权利要求1的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型,所述化合物具有式I-B所示结构,
    Figure PCTCN2016110007-appb-100012
    其中,所述式I-B化合物的限制性条件为,
    当A和B与相连的碳原子形成未取代的苯环,R1、R2和R3均为H,R4为F时,
    1)当式I-B中
    Figure PCTCN2016110007-appb-100013
    Figure PCTCN2016110007-appb-100014
    时,R6不存在,R5不为H、羟基、甲基、异丙基、环丙基、三氟甲基、二氟甲基和三氟乙基;
    2)当式I-B中
    Figure PCTCN2016110007-appb-100015
    Figure PCTCN2016110007-appb-100016
    时,R5和R6不全为H、且当R5或R6其中一个为H时,另一个不为三氟甲基和乙氧羰基;
    3)式I-B中
    Figure PCTCN2016110007-appb-100017
    不为
    Figure PCTCN2016110007-appb-100018
    4)所述环X不为吡咯烷、2-吡咯烷酮、吗啉、3-吗啉酮和2-噁唑烷酮;并且,
    5)所述式I-B化合物不为4-{[3-(3,4-二氢-1H-吡咯并[1,2-a]吡嗪-2-羰基)-4-氟-苯基]甲基}-2H-酞嗪-1-酮、4-{[3-(6,8-二氢-5H-[1,2,4]三唑并[1,5-a]吡嗪-7-羰基)-4-氟-苯基]甲基}-2H-酞嗪-1-酮、4-{[4-氟-3-(2-三氟甲基-6,8-二氢-5H-[1,2,4]三唑并[1,5-a]吡嗪-7-羰基)-苯基]甲基}-2H-酞嗪-1-酮、和4-[4-氟-3-(5,6-二氢-8H-咪唑并[1,5-a]吡嗪-7-羰基)苄基]-2H-酞嗪-1-酮;
    其余各原子或取代基定义如权利要求1所述。
  6. 权利要求5的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型,其中所述式I-B中环X为五元杂芳环且杂原子为氮、或六元含氮脂杂环,优选地,所述
    Figure PCTCN2016110007-appb-100019
    选自以下结构:
    Figure PCTCN2016110007-appb-100020
  7. 权利要求1-6任一项的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型,
    其中,所述R5选自氢、氧基、卤素、C1-10烷基、3-20元环烷基、3-20元脂杂环基、5-10元杂芳基、RO-、RC(O)-、ROC(O)-、RR’N-、RR’NC(O)-、RS(O)a-和RR’NSO2-;任选地,其中所述C1-10烷基、3-20元环烷基、3-20元脂杂环基或5-10元杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素(优选为氟)、羟基、氨基、氰基、 C1-6烷基、C1-6烷氧基、3-8元环烷基、3-8元脂杂环基、6-10元芳基和或5-10元杂芳基的取代基取代,其中a为0、1或2;或者,
    R5并在与之相连的环X上形成多环系统,其中,R5选自脂杂环(例如5-8元脂杂环)和杂芳环(例如5-10元杂芳环);优选地,所述脂杂环或杂芳环为五元或六元含氮脂杂环基或杂芳环;例如为吡咯烷环、吡咯环、咪唑环、吡唑环、三氮唑环、噁唑环、噻唑环、哌啶环、哌嗪环、吡嗪环、吗啉环、硫吗啉环、吡啶环、嘧啶环或哒嗪环;
    R和R’各自独立地选自氢、C1-6烷基、3-8元环烷基、5-8元脂杂环基、6-10元芳基和5-10元杂芳基;任选地,其中所述C1-6烷基、3-8元环烷基、5-8元脂杂环基、6-10元芳基或5-10元杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、C1-4烷基、卤代C1-4烷基、羟基C1-4烷基、RS(O)2-、C1-4烷氧基、3-8元环烷基、5-8元脂杂环基、6-10元芳基和5-10元杂芳基的取代基取代,或者,R和R’与所连的N原子一起形成4-10元脂杂环基;
    优选地,R5选自氢、氧基、卤素、C1-6烷基、3-8元环烷基、3-8元脂杂环基、5-6元杂芳基、RO-、RC(O)-、ROC(O)-、RR’N-、RR’NC(O)-、RS(O)a-和RR’NSO2-;任选地,其中所述C1-6烷基、3-8元环烷基、3-8元脂杂环基、5-6元杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素(优选为氟)、羟基、氨基、氰基、C1-4烷基、C1-4烷氧基和3-6元环烷基的取代基取代,其中a为0、1或2;
    其中,R和R’各自独立地选自氢、C1-4烷基和3-6元环烷基;任选地,其中所述C1-4烷基和3-6元环烷基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、C1-2烷基、C1-2烷氧基和C1-2烷基磺酰基的取代基取代;
    优选地,R5选自氢、氧基、卤素、C1-4烷基、3-6元环烷基、3-6元脂杂环基、5-6元杂芳基、RO-、RC(O)-、ROC(O)-、RR’N-、RR’NC(O)-、RS(O)a-和RR’NSO2-;任选地,其中所述C1-4烷基、3-6元环烷基、3-6元脂杂环基、5-6元杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素(优选为氟)、羟基、氨基、氰基、C1-2烷基、C1-2烷氧基和3-6元环烷基的取代基取代,其中a为0、1或2;
    其中,R和R’各自独立地选自氢、C1-2烷基和3-6元环烷基;任选地,其中所述C1-2烷基和3-6元环烷基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、C1-2烷基、C1-2烷氧基和C1-2烷基磺酰基的取代基取代;
    优选地,R5选自氢、溴、羟甲基、1-羟基乙基、2-羟基-2-丙基、二氟甲基、三氟甲基、1,1-二氟乙基、五氟乙基、环丙基-二氟甲基、1-氨基乙基、环丙基甲基、环丙基、1-甲基4-吡唑基、1-甲基-3-吡唑基、2-甲基-[1.3.4]氧代二唑-5-基、3-甲基-[1.2.4] 氧代二唑-5-基、甲氧基、环丙基氧基、乙酰基、乙氧羰基、环丙基氨基、氨酰基、甲氨酰基、N,N-二甲胺酰基、乙氨酰基、2-羟基乙基氨酰基、2-甲氧基乙基氨酰基、2-羟基异丁氨酰基、2-甲磺酰基乙基氨酰基、甲硫基和甲磺酰基。
  8. 权利要求1-7任一项的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型,其中,所述R6不存在或选自氢、氧基、卤素、C1-10烷基、3-20元环烷基、3-20元脂杂环基、5-10元杂芳基、RO-、RC(O)-、ROC(O)-、RR’N-、RR’NC(O)-、RS(O)a-和RR’NSO2-;任选地,其中所述C1-10烷基、3-20元环烷基、3-20元脂杂环基或5-10元杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素(优选为氟)、羟基、氨基、氰基、C1-6烷基、C1-6烷氧基、3-8元环烷基、3-8元脂杂环基、6-10元芳基和5-10元杂芳基的取代基取代,其中a为0、1或2;
    R和R’各自独立地选自氢、C1-6烷基、3-8元环烷基、5-8元脂杂环基、6-10元芳基和5-10元杂芳基;任选地,其中所述C1-6烷基、3-8元环烷基、5-8元脂杂环基、6-10元芳基或5-10元杂芳基各自独立地被一个或多个(例如1、2、3或4个)选自卤素、羟基、氨基、氰基、C1-4烷基、卤代C1-4烷基、羟基C1-4烷基、RS(O)2-、C1-4烷氧基、3-8元环烷基、5-8元脂杂环基、6-10元芳基和5-10元杂芳基的取代基取代,或者,R和R’与所连的N原子一起形成4-10元脂杂环基;
    优选地,R6不存在或选自氢和卤代C1-10烷基(例如卤代C1-6烷基;例如卤代C1-4烷基;例如三氟甲基);
    优选地,R6不存在或选自氢和三氟甲基。
  9. 权利要求1-8任一项的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型,其中A和B与相连的原子一起形成芳环;优选为苯环。
  10. 权利要求1-9任一项的化合物,其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型,其中R4为卤素;优选为氟。
  11. 权利要求1-10任一项的化合物、其前体药物、代谢物形式、药学上可接受的盐 或酯、或前述的异构体、水合物、溶剂合物或晶型,其中R1、R2和R3为氢原子。
  12. 权利要求1的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型,其中所述化合物选自,
    4-[4-氟-3-(2,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)苄基]-2H-酞嗪-1-酮(化合物1);
    4-[4-氟-3-(2,4,6,7-四氢-吡唑并[4,3-c]吡啶-5-羰基)苄基]-2H-酞嗪-1-酮(化合物2);
    4-[4-氟-3-(6,7-二氢-4H-[1,2,3]三氮唑并[1,5-a]吡嗪-5-羰基)苄基]-2H-酞嗪-1-酮(化合物3);
    N-甲基-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-2-甲酰胺(化合物4);
    N-甲基-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺(化合物5);
    4-[3-(2-环丙甲酰-2,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物6);
    4-[3-(2-环丙基甲基-2,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物7);
    4-[4-氟-3-(2-甲基-2,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)-苄基]-2H-酞嗪-1-酮(化合物8);
    4-[3-(2-环丙甲酰基-2,4,6,7-四氢-吡唑并[4,3-c]吡啶-5-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物9);
    顺-4-[4-氟-3-(4,5,5a,6,7,8,9,9a-八氢-[1,2,4]三氮唑并[4,3-a][1,6]萘啶-7-羰基)-苄基]-2H-酞嗪-1-酮(化合物10);
    反-4-[4-氟-3-(4,5,5a,6,7,8,9,9a-八氢-[1,2,4]三氮唑并[4,3-a][1,6]萘啶-7-羰基)-苄基]-2H-酞嗪-1-酮(化合物11);
    4-[3-(1-环丙基-1,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物12);
    7-[2-氟-5-(4-氧代-3,4-二氢-1-酞嗪基)甲基-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺(化合物13);
    N,N-二甲基-7-[2-氟-5-(4-氧代-3,4-二氢-1-酞嗪基)甲基-苯甲酰基]-5,6-二氢 -8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺(化合物14);
    7-[2-氟-5-(4-氧代-3,4-二氢-1-酞嗪基甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-羧酸乙酯(化合物15);
    4-[4-氟-3-(3-三氟甲基-1,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)苄基]-2H-酞嗪-1-酮(化合物16);
    N-甲基-7-[2-氟-5-(4-氧代-3,4-二氢-1-酞嗪基)甲基苯甲酰基]-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-3-甲酰胺(化合物17);
    7-[2-氟-5-(4-氧杂-3,4-二氢-1-酞嗪基)甲基苯甲酰基]-2-三氟甲基-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-3-羧酸乙酯(化合物18);
    4-[3-(2-环丙基-2,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)-4-氟苯基]-2H-酞嗪-1-酮(化合物19);
    N-甲基-6-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)苯甲酰基]-4,5,6,7-四氢-2H-吡唑[3,4-c]吡啶-3-甲酰胺(化合物20);
    6-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-4,5,6,7-四氢-1H-吡唑[3,4-c]吡啶-3-甲酰胺(化合物21);
    4-[4-氟-3-(2-羟甲基-6,7-二氢-4H-吡唑[1,5-a]吡嗪-5-羰基)-苄基]-2H-酞嗪-1-酮(化合物22);
    4-[3-(3-溴-6,7-二氢-4H-[1,2,3]三氮唑并[1,5-a]吡嗪-5-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物23);
    N-(2-羟乙基)-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺(化合物24);
    N,N-二甲基-6-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-4,5,6,7-四氢-1H-吡唑并[3,4-c]吡啶-3-甲酰胺(化合物25);
    4-[4-氟-3-(6-氧代-1,3,4,6-四氢-吡啶并[1,2-a]吡嗪-2-羰基)苄基]-2H-酞嗪-1-酮(化合物26);
    4-[3-(3-溴-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物27);
    4-[3-(3-溴-6,7,8,9-四氢-5H-[1,2,4]三氮唑并[4,3-d][1,4]二氮杂卓-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物28);
    4-{4-氟-3-[3-(1-羟基-1-甲基-乙基)-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮(化合物29);
    N-甲基-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-2-三氟甲基-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-3-甲酰胺(化合物30);
    N-乙基-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-2-三氟甲基-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-3-甲酰胺(化合物31);
    4-{4-氟-3-[3-(1-羟基-1-甲基-乙基)-2-三氟甲基-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮(化合物32);
    4-[4-氟-3-(3-甲氧基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-苄基]-2H-酞嗪-1-酮(化合物33);
    N-(2-甲氧基乙基)-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺(化合物34);
    N-(2-羟基-2-甲基丙基)-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺(化合物35);
    N-(2-甲基磺酰基乙基)-7-[2-氟-5-(4-氧代-3,4-二氢-酞嗪-1-甲基)-苯甲酰基]-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-3-甲酰胺(化合物36)。
    4-[3-(3-乙酰基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物37);
    4-{4-氟-3-[3-(1-甲基-1H-吡唑-4-基)-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮(化合物38);
    4-{4-氟-3-[3-(1-甲基-1H-吡唑-4-基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮(化合物39);
    4-[4-氟-3-(3-甲巯基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-苄基]-2H-酞嗪-1-酮(化合物40);
    4-[4-氟-3-(3-甲基磺酰基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-苄基]-2H-酞嗪-1-酮(化合物41);
    4-{4-氟-3-[3-(2-羟基丙基-2-基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮(化合物42);
    4-{4-氟-3-[3-(2-羟基丙基-2-基)-1,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基]-苄基}-2H- 酞嗪-1-酮(化合物43);
    4-{4-氟-3-[3-(1-羟基乙基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮(化合物44);
    4-{4-氟-[(3-(1-甲基-1H-吡唑-3-基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮(化合物45);
    4-{3-[3-(1,1-二氟乙基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基]-4-氟苄基}-2H-酞嗪-1-酮(化合物46);
    4-[3-(3-环丙基-1,4,5,7-四氢-吡唑并[3,4-c]吡啶-6-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物47);
    4-[3-(3-环丙基-5,6-二氢-8H-咪唑并[1,5-a]吡嗪-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物48);
    4-[3-(3-环丙基-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物49);
    4-[3-(3-环丙基-2-三氟甲基-5,6二氢-8H-咪唑并[1,2-a]吡嗪-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物50);
    4-{4-氟-3-[3-(3-甲基-[1,2,4]-噁二唑-5-基)-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮(化合物51);
    4-{4-氟-3-[3-(5-甲基-[1,3,4]-噁二唑-2-基)-5,6-二氢-8H-咪唑并[1,2-a]吡嗪-7-羰基]-苄基}-2H-酞嗪-1-酮(化合物52);
    4-[3-(3-环丙基氨基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物53);
    4-[3-(3-环丙基甲基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物54);
    4-{3-[3-(环丙基二氟甲基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物55);
    4-[3-(3-环丙基-6,7-二氢-4H-[1,2,3]三氮唑并[1,5-a]吡嗪-5-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物56);
    4-[3-(3-环丙氧基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物57);
    4-{3-[3-(1-氨基乙基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基]-4-氟苄基}-2H-酞嗪-1-酮(化合物58);
    4-[3-(3-环丙基-1-三氟甲基-5,6-二氢-8H-咪唑并[1,5-a]吡嗪-7-羰基)-4-氟苄基]-2H-酞嗪-1-酮(化合物59);
    4-{3-[3-(1,1-二氟乙基)-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基]-4-氟苄基}-7-氟-2H-酞嗪-1-酮(化合物60);
    4-[3-(3-环丙基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-4-氟苄基]-7-氟-2H-酞嗪-1-酮(化合物61);
    4-[3-(3-二氟甲基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)-4-氟苄基]-7-氟-2H-酞嗪-1-酮(化合物62);
    4-[4-氟-3-(3-全氟乙基-5,6-二氢-8H-[1,2,4]三氮唑并[4,3-a]吡嗪-7-羰基)苄基]-2H-酞嗪-1-酮(化合物63);
    7-氟-4-[4-氟-3-(3-五氟乙基-5,6,7,8-四氢-[1,2,4]三唑[4,3-a]吡嗪-7-羰基)苄基]-2H-酞嗪-1-酮(化合物64);和
    4-[4-氟-3-(6-甲基-3-五氟乙基-5,6,7,8-四氢-[1,2,4]三唑[4,3-a]吡嗪-7-羰基)苄基]-2H-酞嗪-1-酮(化合物65)。
  13. 权利要求1的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型的制备方法,其选自以下方法:
    (1)当式I中R5为RR’NC(O)-时,所述化合物可通过以下路线合成:
    Figure PCTCN2016110007-appb-100021
    将化合物A与化合物B’进行缩合反应得到化合物C;将化合物C与化合物RR’NH 进行氨酯交换得到化合物D;
    (2)当式I中R5为RC(O)-时,所述化合物可通过以下路线合成:
    Figure PCTCN2016110007-appb-100022
    将化合物A与化合物B”进行缩合反应得到化合物C’;将化合物C’与化合物RCO-Lg进行酰基化反应得到化合物D’;或者,
    (3)当式I中R5和R6为权利要求1中所述的其它基团时,所述化合物可通过以下路线合成:
    Figure PCTCN2016110007-appb-100023
    将化合物A与化合物B进行缩合反应得到式I化合物;
    其中,RCO-Lg中的Lg表示离去基团,例如卤素、-OTs、-OCOR等,其余各原子和取代基的定义如权利要求1所述。
  14. 药物组合物,其包含权利要求1-12任一项所述的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型;任选地,其还包含药学可接受的载体或赋形剂。
  15. 权利要求14所述的药物组合物,其中所述药物组合物含有0.01-2000mg;优选为0.1-1000mg;更优选为1-800mg;更优选为10-600mg;特别优选为50-500mg的权利要求1-12任一项所述的化合物、其前体药物、代谢物形式、药学上可接受的盐 或酯、或前述的异构体、水合物、溶剂合物或晶型。
  16. 权利要求14或15的药物组合物,其还包含一种或多种抗肿瘤药物;优选地,所述抗肿瘤药物选自替莫唑胺、阿霉素、紫杉醇、顺铂、卡铂、达卡巴嗪、拓扑替康、伊立替康、吉西他滨、贝伐单抗、anti-CTLA-4单抗Ipilimumab、anti-PD-1单抗pembrolizumab和Nivolumab以及anti-PD-L1单抗atezolizumab。
  17. 权利要求1-12任一项所述的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型或权利要求14-16任一项所述的药物组合物在制备PARP抑制剂中的用途;优选地,其中所述PARP抑制剂为PARP-1抑制剂。
  18. 权利要求1-12任一项所述的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型或权利要求14-16任一项所述的药物组合物在制备治疗肿瘤的辅助剂或是用于增强放射或化学治疗肿瘤效果的药物中的用途。
  19. 权利要求1-12任一项所述的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型或权利要求14-16任一项所述的药物组合物在制备治疗肿瘤的药物中的用途。
  20. 权利要求1-12任一项所述的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型或权利要求14-16任一项所述的药物组合物在制备治疗血管疾病、神经退变性疾病或神经系统炎症药物中的用途。
  21. 一种抑制PARP活性的方法,所述方法包括向受试者施用有效量的权利要求1-12任一项所述的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型或权利要求14-16任一项所述的药物组合物;优选地,其中所述PARP为PARP-1。
  22. 一种辅助治疗肿瘤或增强放射或化学治疗效果的方法,所述方法包括向受试者施用有效量的权利要求1-12任一项所述的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型或权利要求14-16任一项所述的药物组合物。
  23. 一种治疗肿瘤的方法,所述方法包括向受试者提供有效量的权利要求1-12任一项所述的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型或权利要求14-16任一项所述的药物组合物。
  24. 一种治疗血管疾病、神经退变性疾病或神经系统炎症疾病的方法,所述方法包括向受试者施用有效量的权利要求1-12任一项所述的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型或混合物或权利要求14-16任一项所述的药物组合物。
  25. 权利要求1-12任一项所述的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型或权利要求14-16任一项所述的药物组合物,其用于抑制PARP活性;优选地,其其用于抑制PARP-1活性。
  26. 权利要求1-12任一项所述的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型或权利要求14-16任一项所述的药物组合物,其用于辅助治疗肿瘤或用于增强放射或化学治疗肿瘤效果。
  27. 权利要求1-12任一项所述的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型或权利要求14-16任一项所述的药物组合物,其用于治疗肿瘤。
  28. 权利要求1-12任一项所述的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型或权利要求14-16任一项所 述的药物组合物,其用于治疗血管疾病、神经退变性疾病或神经系统炎症。
  29. 权利要求18-19任一项的用途、权利要求22-23任一项的方法或权利要求26-27所述的化合物、其前体药物、代谢物形式、药学上可接受的盐或酯、或前述的异构体、水合物、溶剂合物或晶型,其中所述肿瘤选自乳腺癌、卵巢癌、结直肠癌、黑色素瘤、肺癌、胃肠道间质瘤、脑癌、宫颈癌、胰腺癌、前列腺癌、胃癌、慢性髓样白细胞过多症、肝癌、淋巴瘤、腹膜癌、软组织肉瘤、神经内分泌肿瘤和胶质母细胞瘤。
PCT/CN2016/110007 2015-12-16 2016-12-15 酞嗪酮衍生物、其制备方法及用途 WO2017101796A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201680007644.0A CN107207504B (zh) 2015-12-16 2016-12-15 酞嗪酮衍生物、其制备方法及用途

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201510939891.1 2015-12-16
CN201510939891 2015-12-16

Publications (1)

Publication Number Publication Date
WO2017101796A1 true WO2017101796A1 (zh) 2017-06-22

Family

ID=59055859

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2016/110007 WO2017101796A1 (zh) 2015-12-16 2016-12-15 酞嗪酮衍生物、其制备方法及用途

Country Status (2)

Country Link
CN (1) CN107207504B (zh)
WO (1) WO2017101796A1 (zh)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109251204A (zh) * 2017-07-13 2019-01-22 中国药科大学 含有酞嗪-1(2h)-酮结构的parp抑制剂、其制法及医药用途
CN109748923A (zh) * 2019-01-31 2019-05-14 中国药科大学 含苯并[4,5]咪唑[1,2-a]吡嗪酮类衍生物及其制备方法与用途
CN112724148A (zh) * 2020-12-29 2021-04-30 上海药明康德新药开发有限公司 一种咪唑并哒嗪类化合物及修饰后的双亲功能分子及其应用
CN113087724A (zh) * 2020-01-08 2021-07-09 四川科伦博泰生物医药股份有限公司 异噻唑并嘧啶酮类化合物,包含其的药物组合物及其用途
WO2023288002A1 (en) * 2021-07-16 2023-01-19 Oregon Health & Science University Phthalazinone-based parp-1 inhibitors

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012019427A1 (zh) * 2010-08-09 2012-02-16 上海恒瑞医药有限公司 酞嗪酮类衍生物、其制备方法及其在医药上的应用
WO2012019426A1 (zh) * 2010-08-09 2012-02-16 上海恒瑞医药有限公司 酞嗪酮类衍生物、其制备方法及其在医药上的应用
WO2012072033A1 (zh) * 2010-12-03 2012-06-07 苏州科瑞戈医药科技有限公司 取代的2,3-二氮杂萘酮化合物及其用途
CN102898377A (zh) * 2012-02-14 2013-01-30 南京圣和药业有限公司 一类新型酞嗪酮衍生物及其用途
WO2014019468A1 (zh) * 2012-08-01 2014-02-06 中国科学院上海药物研究所 哌嗪并三唑类化合物及其制备方法和制药用途
WO2014102817A1 (en) * 2012-12-31 2014-07-03 Cadila Healthcare Limited Substituted phthalazin-1 (2h)-one derivatives as selective inhibitors of poly (adp-ribose) polymerase-1

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012019427A1 (zh) * 2010-08-09 2012-02-16 上海恒瑞医药有限公司 酞嗪酮类衍生物、其制备方法及其在医药上的应用
WO2012019426A1 (zh) * 2010-08-09 2012-02-16 上海恒瑞医药有限公司 酞嗪酮类衍生物、其制备方法及其在医药上的应用
WO2012072033A1 (zh) * 2010-12-03 2012-06-07 苏州科瑞戈医药科技有限公司 取代的2,3-二氮杂萘酮化合物及其用途
CN102898377A (zh) * 2012-02-14 2013-01-30 南京圣和药业有限公司 一类新型酞嗪酮衍生物及其用途
WO2014019468A1 (zh) * 2012-08-01 2014-02-06 中国科学院上海药物研究所 哌嗪并三唑类化合物及其制备方法和制药用途
WO2014102817A1 (en) * 2012-12-31 2014-07-03 Cadila Healthcare Limited Substituted phthalazin-1 (2h)-one derivatives as selective inhibitors of poly (adp-ribose) polymerase-1

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109251204A (zh) * 2017-07-13 2019-01-22 中国药科大学 含有酞嗪-1(2h)-酮结构的parp抑制剂、其制法及医药用途
CN109748923A (zh) * 2019-01-31 2019-05-14 中国药科大学 含苯并[4,5]咪唑[1,2-a]吡嗪酮类衍生物及其制备方法与用途
CN113087724A (zh) * 2020-01-08 2021-07-09 四川科伦博泰生物医药股份有限公司 异噻唑并嘧啶酮类化合物,包含其的药物组合物及其用途
CN112724148A (zh) * 2020-12-29 2021-04-30 上海药明康德新药开发有限公司 一种咪唑并哒嗪类化合物及修饰后的双亲功能分子及其应用
WO2023288002A1 (en) * 2021-07-16 2023-01-19 Oregon Health & Science University Phthalazinone-based parp-1 inhibitors

Also Published As

Publication number Publication date
CN107207504B (zh) 2020-03-10
CN107207504A (zh) 2017-09-26

Similar Documents

Publication Publication Date Title
EP3377482B1 (en) Modulators of ror-gamma
WO2019158019A1 (zh) 嘧啶并环化合物及其制备方法和应用
WO2018157857A1 (zh) 凋亡信号调节激酶抑制剂及其制备方法和应用
WO2017101796A1 (zh) 酞嗪酮衍生物、其制备方法及用途
JP2020519589A (ja) G12c変異型rasタンパク質を阻害するヘテロアリール化合物
JP6564406B2 (ja) カゼインキナーゼ1デルタ/イプシロン阻害剤としてのイミダゾ−ピリダジン誘導体
WO2018045956A1 (zh) 苯并咪唑类化合物激酶抑制剂及其制备方法和应用
EP3035800A1 (en) Compounds inhibiting leucine-rich repeat kinase enzyme activity
KR20220100879A (ko) Fgfr 저해제로서의 이환식 헤테로사이클
CN112165944B (zh) 转录激活蛋白的咪唑并哌嗪抑制剂
WO2022194245A1 (zh) 嘧啶并环类化合物及其制法和用途
AU2014400628A1 (en) Aminopyridazinone compounds as protein kinase inhibitors
KR20140096033A (ko) 신규 화합물
EP2815750A1 (en) 5-cyano-4-(pyrrolo [2,3b] pyridine-3-yl)-pyrimidine derivatives useful as protein kinase inhibitors
BR112014017749B1 (pt) Composto alquinilbenzeno 3,5-dissubstituído e sal do mesmo
TW201333004A (zh) 新穎化合物
WO2018077246A1 (zh) 用作神经营养因子酪氨酸激酶受体抑制剂的氨基吡唑并嘧啶化合物
CA2831346A1 (en) Bicyclic heterocycle compounds and their uses in therapy
WO2019120234A2 (zh) 作为溴结构域蛋白质抑制剂的化合物和组合物
CA2849751A1 (en) Chiral n-acyl-5,6,7,(8-substituted)-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazines as selective nk-3 receptor antagonists, pharmaceutical composition, methods for use in nk-3 receptormediated disorders and chiral synthesis thereof
EP3638680B1 (en) Heteroaromatic compounds as vanin inhibitors
WO2016192630A1 (zh) 一类具有激酶抑制活性的化合物、制备方法和用途
WO2022160931A1 (zh) 吡啶并嘧啶类衍生物及其制备方法和用途
CN111032643A (zh) 新的喹啉酮化合物
AU2019280356B2 (en) ERK inhibitor and use thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 16874853

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16874853

Country of ref document: EP

Kind code of ref document: A1